WO2014210454A1 - Treatment and diagnosis of ocular disease - Google Patents

Treatment and diagnosis of ocular disease Download PDF

Info

Publication number
WO2014210454A1
WO2014210454A1 PCT/US2014/044567 US2014044567W WO2014210454A1 WO 2014210454 A1 WO2014210454 A1 WO 2014210454A1 US 2014044567 W US2014044567 W US 2014044567W WO 2014210454 A1 WO2014210454 A1 WO 2014210454A1
Authority
WO
WIPO (PCT)
Prior art keywords
ocular
ltxa
pharmaceutical composition
eye
subject
Prior art date
Application number
PCT/US2014/044567
Other languages
French (fr)
Inventor
Scott KACHLANY
Benjamin BELINKA
Original Assignee
Rutgers, The State University Of New Jersey
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Rutgers, The State University Of New Jersey filed Critical Rutgers, The State University Of New Jersey
Priority to JP2016524239A priority Critical patent/JP6544806B2/en
Priority to US14/901,631 priority patent/US9950030B2/en
Priority to CA2917056A priority patent/CA2917056C/en
Priority to EP14817083.0A priority patent/EP3013425B1/en
Publication of WO2014210454A1 publication Critical patent/WO2014210454A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/164Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0048Eye, e.g. artificial tears
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0048Eye, e.g. artificial tears
    • A61K9/0051Ocular inserts, ocular implants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection

Definitions

  • This invention relates to pharmaceutical compositions, reagents and methods for treating or diagnosing ocular disease.
  • Ocular inflammatory disease is a general term embracing a number of diseases and conditions in whic inflammation affects the eye or surrounding tissues. Inflammation is due to abnormal activation and migration of white blood cells to regions for example., the conjunctival epithelium and tear glands. Although some treatments are presently available, many possess significant side effects or are not very effective in alleviating all symptoms. Thus, there is a need for drugs and reagents for treatment and diagnosis of ocular inflammatory disease.
  • This invention relates to treatment and diagnosis of ocular inflammatory disease using leukotoxin (LfxA), a bacterial protein.
  • At least one aspect of the invention is directed to a method of treating a disease characterized by ocular inflammation, the method comprising administering a pharmaceutical composition to a subject in need of such treatment i an amount effective to reduce said inflammation, wherein the pharmaceutical composition comprises a leukotoxin and a pharmaceutically acceptable carrier, and wherein the disease is an ocular inflammatory disease.
  • the ocular inflammatory disease maybe keratoconjuuctivitis sicca, diabetic retinopath ..
  • diabetic macula edema Sjogren syndrome, dry eye, scleritis, birdshot retinochoriodopathy, ocular cicatricial pemphigoidm, keratitis, sympathetic ophthalmia, vogt-koyanagi harada, fuehs' helerochromk iridocyclitis, uveitis, pars planitis, episcleritis, optic neuritis, orbital pseudotumor, retinal vasculitis, ocular allergy, and chronic conjunctivitis.
  • Another aspect of the invention is directed to method of reducing ocular inflammation in an ocular regio within a subject in need thereof, characterized by increased levels of activated white blood cells in a subject in need thereof, the method comprising administering to the subject an amount of a pharmaceutical composition in amount effective to reduce said ocular inflammation, wherein the pharmaceutical composition comprises a leukotoxin and a pharmaceutically acceptable carrier.
  • the ocular inflammation may be caused by an infection., autoimmune disease, allergen, ocular trauma, and ocular allograft rejection.
  • the infection may be a viral, bacterial, or a fungal infection.
  • the leukotoxin is LtxA, prepared from Aggregatibacter actinomyceiemcomitans.
  • LtxA has ai least 90% homology with the peptide according to SEQ ID NO: ⁇ .
  • the pharmaceutical composition comprising leukotoxin is administered topically to the ocular region.
  • the pharmaceutical composition is in a form suitable for topical administration.
  • the pharmaceutical composition is formulated for and administered by using an eye drop dispenser, in another embodiment, the pharmaceutically acceptable carrier is an eye gel, eye cream, suspension-type eye drops, eye wash, ointment, gel, liposomal dispersion, colloidal microparticle suspension, implant or a contact lens.
  • In another embodiment the anio tit of the pharmaceutical composition comprising leukotoxin administered to a subject in need thereof is effective to reduce local cytokine levels in the ocular region. One or more of the following cytokines expression levels are reduced, iL-4, IL ⁇ 5 JL ⁇ 9 ; ILHF and IL ⁇ 23a.
  • kits comprising a pharmaceutically effective amount of leukotoxin for the treatment of an ocular inflammatory disease.
  • the kit may contain multiple doses of a pharmaceutical composition comprising m effective amount of leukotoxin to treat an ocular inflammatory disease or to reduce ocular inftammation characterized by the increase of activated white blood ceils in the ocular region that express a greater level of LFA- 1 compared to white blood cells from a normal healthy subject.
  • FIG. I is a diagram showing migration of white blood ceils (WBCs) across an endothel ial barrier.
  • FIG. 2 is a diagram showing sensitivity of activated T-ceils
  • FIG. 3A-F is a set of diagrams showing the activity of LtxA under physiologic conditions.
  • FIG. 4 is a diagram showing the examination of cytokines in Sung tissue following treatment of house dust mite (HDM) exposed mice with either a vehicle (saline, HDM/Dex vehicle; or HDM/LtxA vehicle); dexaroethasone (HDM/Dex); or leukotoxin (HDM/LtxA), DETAILED DESCRIPTION OF THE INVENTIO
  • a vehicle saline, HDM/Dex vehicle; or HDM/LtxA vehicle
  • dexaroethasone HDM/Dex
  • HDM/LtxA leukotoxin
  • This invention relates to reagents and methods for treating or diagnosing ocular inflammatory disease and incorporates the discovery that administering LxA to a subject sufferin ocular inflammation characterized by activaied inflammatory cells expressing LFA-1 results in the depletion of the activated inflammatory cells.
  • LtxA targets inflammatory white blood cells WBCs thai express the activated conformation of LFA-1 o their surface that migrate to the eye and surrounding tissue resulting i ocular inflammation and local organ damage.
  • Ocular inflammatory disease is a general term for inflammation affecting the ocular region.
  • the ocular region means any part of the eye or surrounding tissue, as known by one with ordinary skill in the art. Inflammation involving the eye can range from the familiar allergic conjunctivitis of hay fever to rare, potentially blinding conditions such as uveitis, scleiitis, episcleritis, optic neuritis, keratitis, orbital pseudotumor, retinal vasculitis, and chronic conjunctivitis.
  • fOOISJ fOOISJ
  • ocular inflammatory disease if inflammation develops in the eye(s), or in the optic nerve, blood vessels, muscles or other tissues that surround the eye, the resulting illness is referred to as an ocular inflammatory disease.
  • the location of the inflammation may govern the diagnostic name for the ocular inflammatory disease.
  • uveitis inflammation in the uveal tract
  • sc!eritis inflammation of the sclera
  • pars planitis inflammation of the pars plana, and so forth.
  • the treatment of ocular inflammatory disease is aimed at .reducing inflammation.
  • Ocular inflammation can occur for many reasons for example, as a resul t of an infec tion, an autoimmune disorder, trauma to the eye, or allergies.
  • LtxA is a - 1 15 kDa protein produced by the Gram negative bacterium Aggregatibacter actinomycetemeomitans (Kac any, S. C. 2010. J Dent Res 89:563-570.). LtxA specifically kills leukocytes of humans and Old World Primates by forming pores in the membrane and causing apoptosis or necrosis (Mangan et a!., 1991. infect Immim 59:3267-72.). LtxA binds specifically to LFA ⁇ i and cells that lack LFA-1 are resistant to its toxicity (KachJany, S. C. et al., 2010.
  • LtxA is not. active against human red. blood ceils, human epithelial cells, rat cells, or mou.se cells. LtxA also remains active in the presence of human peripheral blood.
  • LtxA preparations can be used, highly purified LtxA is preferred.
  • LtxA polypeptide purified from Aggregatibacter actinomycetemeomitans SEQ ID NO: 1 shown above
  • other variants having substantially the same bioiogical activity as that having the sequence of SEQ ID NO: 1.
  • Aggregatibacter actinomycetemeomitans secreted active LtxA into culture superaatants (Kaeh ' iany, S. C, et al. 2000. infect Immun 68:6094-100) and an efficient method for its purification was described in Kach!any, S. C, et at 2002.
  • This method can therefore be used to prepare isolated or purified LtxA polypeptide.
  • a purification procedure of the toxin involves;
  • polypeptide refers to polypeptide that has been separated from other proteins, lipids, and nucleic acids with which it is naturally associated.
  • polypeptide and protein can be used interchangeably when referring to LtxA.
  • the polypeptide constitutes at least 10% (i.e., any percentage between 10% and 100%, e.g., 20%, 30%. 40%, 50%, 60%, 70 %, 80%, 85%, 90%, 95%, and 99%) by dry weight of the purified preparation. Purity can be measured by any appropriate standard method, for example, by column chromatography, polyacrylaxnide gel electrophoresis, or HPLC analysis.
  • LtxA refers to a polypeptide derivative of the LtxA polypeptide, e.g., a protein having one or more point mutations, insertions, deletions, truncations, a fusion protein, or a combination thereof. It retains substantially the activity of the LtxA polypeptide, i.e., the ability to .target and kill WBCs that express the activated conformation of LFA ⁇ 1. on their surface while having littie or no toxic effect o» other cells or organs in the body.
  • the isolated polypeptide can contain SEQ I D NO; i or a functional fragment of SEQ ID NO; L
  • the functional equivalent is at least 75% (e.g., any number between 75% and 100%, inclusive, e.g., 70 %, 80%, 85%, 90%, 95%, and 99%) identical to SEQ ID NO: I .
  • LtxA obtained by recombinant NA technology may have the same amino acid sequence as naturally a occurring LtxA (SEQ 3D NO: 1 ) or an functionally equivalent thereof.
  • LtxA also covers chemically modified LtxA. Examples of chemically modified LtxA include LtxA subjected to conformational change, addition or deletion of a sugar chain, and LtxA to which a compound such as polyethylene glycol has been bound. Once purified and tested by standard methods or according to the method described in the examples below. LtxA can be included in a pharmaceutical composition, e.g., a topical composition.
  • amino acid composition of the LtxA polypeptide described herein may vary without disrupting the ability of the polypeptide to target and kill WBCs.
  • it can contain one or more conservative amino acid substitutions.
  • a "conservative amino acid substitution” is one in which the amino acid residue is replaced with an amino acid residue having a similar side chain. Families of amino acid residues having similar side chains have been defined in the art.
  • amino acids with basic side chains e.g., lysine, arginine, histidine
  • acidic side chains e.g., aspartic acid, glutamic acid
  • uncharged polar side chains e.g., glycine, asparagine, glutaniine, serine, threonine, tyrosine, cysteine
  • nonpolar side chains e.g., alanine, valine, leucine, isoleueine, proline, phenylalanine, methionine, tryptophan
  • beta-branched side chains e.g., threonine, valine, isoleucine
  • aromatic side chains e.g...
  • a predicted nonessential amino acid residue in SEQ ID NO; 1 is preferably replaced with another amino acid residue from the same side chain family.
  • mutations can be introduced randomly along all or part of SEQ ID NO: , such as by saturation mutagenesis, and the resultant mutants can be screened for the ability to mprove the ocular condition and/or to identify mutants that retain, the activity as described below in the examples.
  • a LtxA polypeptide as described in this invention can be obtained as a naturally occurring polypeptide or a recombinant polypeptide.
  • a nucleic acid encoding it e.g., SEQ ID NO: 2
  • another nucleic acid encoding a fusion partner e.g., gl «tathione-s-transferase (GST), 6x-His epitope tag, or l 3 Gene 3 protein.
  • the resultant fusion nucleic acid expresses in suitable host cells a fusion protein that can be isolated by .methods known in the art.
  • the isolated fusion protein can be further treated, e.g. , by enzymatic digestion, to remove the fusion partner and obtain the recombinant polypeptide of this invention.
  • the present invention also provides a pharmaceutical composition that contains LtxA and a pharmaceutically acceptable carrier suitable for ocular administration.
  • pharmaceutically acceptable carriers include, but are not limited to eye gel, eye cream, suspension-rype eye drops, eye wash, ointment, gel, liposomal dispersion, colloidal microparticle suspension, contact lens, and the like, and other preparations known in the art to be suitable for ocular administration.
  • the pharmaceutical compositions of the present invention containing LtxA may be admimstered using commonly known devices configured for the delivery of the pharmaceutical compositions in the form of to the region surrounding the eye.
  • An ocular insert may also comprise a biodegradable controlled release polymeric matrix.
  • the ocular insert can be implanted in the conjunctiva, sclera, pars plana, anterior segment, or posterior segment of the eye. Implants provide for controlled release of the pharmaceutical composition to the ocular surface, typically sustain, release of LtxA over an extended time period.
  • the pharmaceutically acceptable carrier of the pharmaceutical composition of the invention may comprise a wide variety of non-active ingredients which are useful for formulation purposes and which do not materially affect the novel and useful properties of LtxA.
  • a “pharmaceutically acceptable” or “ophthalmologically acceptable” component means a component that is not biologically or otherwise undesirable, i.e., the component may be incorporated into a pharmaceutical composition of the invention and administered topically to a patient's eye without causing any undesirable biological effects or interacting in a deleterious manner with any of the other components of the formulation composition in which it. is contained.
  • pharmaceutically acceptable refers to a component other than a pharmacologically active agent, it is implied that the component has met the required standards of toxicological and manufacturing testing or that it is included on the Inactive Ingredient Guide prepared by the U.S. Food and Drug Administration.
  • Suitable thickeners will be known to those of ordinary skill in the art of ophthalmic formulation, and include, by way of example, cellulosic polymers such as methyl cellulose (MC), hydroxyethyketlulose (HEC), hvdroxypropylcellubse ⁇ HPC), hydrox ropylmeth Ice 1.1 nlose (HPMC), and sodium carboxyraethylce!luJose ( aCMC), and other swellabte hydrophilic polymers such as polyvinyl alcohol (PVA), hyaluronic acid or a salt thereof (e.g., sodium hyaluronate), and cross!inked acrylic acid polymers commonly referred to as "carboraers”.
  • PVA polyvinyl alcohol
  • hyaluronic acid or a salt thereof e.g., sodium hyaluronate
  • carboraers cross!inked acrylic acid polymers commonly referred to as "carboraers”.
  • any thickener is such that a viscosity in the range of about 15 cps to 25 cps is provided, as a solution having a viscosity in the aforementioned range is generally considered optimal for both comfort and retention of the formulation in the eye.
  • Any suitable isotonic agents and buffering agents commonly used in ophthalmic formulations may be used, providing that the osmotic pressure of the solution does not deviate from that of lachrimal fluid by more than 2-3% and that the pH of the .formulation is maintained in. the range of about 6.5 to about 8.0, preferably in the range of about 6.8 to about 7.8, and optimally at a pH of about 7.4.
  • Preferred buffering agents include carbonates such as sodium and potassium bicarbonate.
  • compositions of the invention may also be prepared as a hydrogei, dispersion, or colloidal suspension.
  • Hydrogels are formed by incorporation of a swei iahie, gel- forming polymer such as those set forth above as suitable thickening agents (i.e., MC, EEC, HPC, HPMC, NaCMC, PVA, or hyaluronic acid or a salt thereof, e.g., sodium hyaluronate), except that a formulation referred to in the ait as a "hydrogei" ' typically has a higher viscosity than a formulation referred to as a "thickened” solution or suspension.
  • suitable thickening agents i.e., MC, EEC, HPC, HPMC, NaCMC, PVA, or hyaluronic acid or a salt thereof, e.g., sodium hyaluronate
  • a pharmaceutical composition may also be prepared so as to form a hydrogei in situ following application to the eye.
  • Such gels are liquid at room temperature but get at higher temperatures (and thus are termed "thermoreversible” hydrogels), such as when placed in contact with body fluids.
  • Biocompatible polymers thai impart this property include acrylic acid polymers and copolymers, N-isopropylacrylamide derivatives, and ABA block copolymers of ethylene oxide and propylene oxide.
  • the pharmaceutical compositions can also be prepared in the form of a dispersion or colloidal suspension.
  • Preferred dispersions are liposomal, in which case the pharmaceutical composition is enclosed within "liposomes," microscopic vesicles composed of alternating aqueous compartments and lipid biiayers.
  • Colloidal suspensions are generally formed from micropariicles, i.e., from microspheres. nanospheres, microcapsules, or nanocapsules, wherein microspheres and nanospheres are generally monolithic particles of a polymer matrix in which the pharmaceutical composition is trapped, adsorbed, or otherwise contained, while with microcapsules and nanocapsules, the formulation is actually encapsulated.
  • the pharmaceutically acceptable ophthalmic carrier used with the pharmaceutical composition of the invention may be of a wide range of types known to those of skill in the art.
  • the pharmaceutical compositions of the invention can be provided as an ophthalmic solution or suspension, in which case the carrier is at least partially aqueous.
  • the pharmaceutical compositions may also be ointments, in which case the pharmaceutical ly acceptable carrier comprises an ointment base.
  • Preferred ointment bases herein have a melting or softening point close to body temperature, and any ointment bases commonly used in ophthalmic preparations ma be advantageously employed.
  • Common ointment bases include petrolatum and mixtures of petrolatum and mineral oil.
  • the pharmaceutical composition may also be incorporated into a sterile ocular insert tha provides for controlled release of the fonnulation over an extended time period, generally in the range of about 12 hours to 60 days, and possibly up to 12 months or more, following implantation of the insert into the conjunctiva, sclera, or pars plana, or into the anterior segment or posterior segment of the eye.
  • a sterile ocular insert tha provides for controlled release of the fonnulation over an extended time period, generally in the range of about 12 hours to 60 days, and possibly up to 12 months or more, following implantation of the insert into the conjunctiva, sclera, or pars plana, or into the anterior segment or posterior segment of the eye.
  • One type of ocular insert is an implant in the form of a monolithic polymer matrix that, gradually releases the pharmaceutical composition to the eye through diffusion and/or matrix degradation. With such an insert, it is preferred that the polymer be completely soluble and or biodegradable (i.
  • inserts are well known in the art, and are typically composed of a water-swellable, gel-forming polymer such as poly ethylene glycol. collagen, polyvinyl alcohol, or a cellulosic polymer.
  • a water-swellable, gel-forming polymer such as poly ethylene glycol. collagen, polyvinyl alcohol, or a cellulosic polymer.
  • Another type of insert thai can be used to deliver the present formulation is a diffusional implant in which the formulation is contained in a central reservoir enclosed within a permeable polymer membrane that allows for gradual diffusion of the pharmaceutical composition out of the implant.
  • Osmotic inserts may also he used, i.e., implants in which the pharmaceutical composition is released as a result of an increase in osmotic pressure within the implant following application to the eye and subsequent absorption of tears.
  • controlled release refers to an agent-containing foundation or fraction thereof in which release of the agent is not immediate, i.e., with a “controlled release” formulation, administration does not result in immediate release of the agent into an absorption pool
  • controlled release refers to "sustained release” rather than to "delayed release” formulations.
  • the terra “sustained release” (synonymous with “extended release”) is used in its conventional sense to refer to a formulation that provides for gradual release of an agent over an extended period of time.
  • the LtxA is released over a period of at least 12 hours, at least 18 hours, at least 24 hours, at least 48 hours, at least 3 days, at least 7 days, at least 14 days, at least 30 days, at least 60 days, at least 90 days, at least 100 days, at least 120 days, at least 150 days, at least 180 days, or even longer.
  • the LtxA or pharmaceutical composition can be administered, as described herein, according to any of a number of standard methods including, but not limited to injection, nasal spray, time-release implant, transdermal patch, eye drops, gels, ointments, orally, intraocular injection, subeonjuetival injection, peri-Zreirobulbar injection, transdermal ⁇ , or topically to tlie ocular region by an eye drop dispenser, or the like, including topical intranasal administration or administration by inhalant, and the like, spray, emulsion, suspension, via any drug carriers as sponges, contact lenses, polymers, microspheres, and implants.
  • the topical administration can he ophihalmically or intraaasa!ly.
  • Topical ophthalmic products may be packaged in multidose form.
  • Preservatives are thus required to prevent microbial contamination during use. Suitable preservatives include: biguanides, hydrogen peroxide, hydrogen peroxide producers, ben alkonium chloride, chlorobutanol, benzododectniu bromide, methyl paraben, propyl parahen, phenylethyl alcohol, edetate disodium, sorbic acid, po!yqiiatemium-3 , or other agents known to those skilled in the art.
  • Such preservatives are typicall employed at a level of from 0.001 to 1 % (w/w). Unit, dose formulations of the present invention will, be sterile, but typically unpreserved. Such formulations, therefore, generally will not contain preservatives.
  • the pharmaceutical composition may further comprise antibiotics.
  • antibiotics include without limitation, cefazolin, cephradine, cefaclor, cephapirin, ceftizoxime, cefoperazone, cefotetan, cefutoxime, cefotaxime, cefadroxil, ceftazidime, cephalexin, cephalothin, cefamandole, cefoxitin, cefoterrorismd, cefbranide, ceftriaxone, cefadroxil, cephradine, cefuroxime, ampicillin, amoxicillin, cyelacillm, ampicillin, penicillin G, penicillin V potassium, piperacillin, oxacillin, bacampicillin, cioxacillin, ticarci!ltn, azlociiUn.
  • the pharmaceutical composition may further comprise corticosteroids.
  • corticosteroids examples include cortisone, prednisolone, triamcinolone, fiaromethokm ⁇ dexameihasone, medrysone, loteprednol, fiuazacort, hydrocortisone, prednisone triamcinolone, betamethasone, prednisone, methyl prednisolone, triamcinolone acetoni.de, triamcinolone hexacatonide, parame hasone aceiate, difiorasone. fluocinolooe and fluoctnonide, derivatives thereof, and mixtures thereof.
  • the pharmaceutical composition may further comprise immunosuppressive agents.
  • immunosuppresive agents include cyclosporine, azathioprine, tacrolimus, T Fa- inhibitors. infliximab, (Remicade), adalimumah Humira), ceriolizumab pegol (Cimzia), and golimumab (Stmponi), and etanereept (Enbrei) and derivatives thereof.
  • the pharmaceutical composition may further comprise antiviral agents.
  • antiviral agents include and are not limited to, interferon gamma, zidovudine, amantadine hydrochloride, ribavirin, acyclovir, valciclovir, dideoxycytidaie, and derivatives thereof.
  • the pharmaceutical composition may further comprise antihistamines.
  • antihistamines include, and are not limited to, loradatine, hydroxyzine, diphenhydramine, chlorpheniramine, brompheniramine, cyproheptadine, terfenadine, clemastine, triprolidine, carbinox amine, diphenylpyr aline, phenindamine, azatadine, tripe lennamine, dexchlorpheniraniine, dexbrompheniramine, methdilaziiie, and trimprazine dox lamine, pheniramine, pyri!amine, chiorcycliztne, thonzylamine, and derivatives thereof
  • the invention provides a method of treating a disease characterized by ocular inflammation in a subject characterized by increased le vels of activated WBCs, by administering to the subject a pharmaceutical composition comprising LtxA and a pharmaceutically acceptable carrier in an amount effective to reduce ocular inflammation characterizing the disease.
  • the invention provides a method of reducing ocular inflammation in a subject characterized by increased levels of activated WBCs, by administering to the subject a pharmaceutical composition comprising leufcotoxm and a pharmaceutically acceptable carrie in an amount effective to reduce ocular inflammation.
  • Ocular inflammatory disease is characterized by a number of diseases and conditions in which inflammation affects the eye or surrounding tissues.
  • ocular inflammatory disease include keratoconjunctivitis sicca, diabetic retinopathy, diabetic macula edema, Sjogren syndrome, dry eye, scleritis, birdshot retinochoriodopathy, ocular cicatricial pemphigoid, keratitis, sympathetic ophthalmia, vogt-koyanagi liarada, fuchs* beterochromic iridocyclitis- uveitis, pars piarsitis, episcleritis, optic neuritis, orbital pseudotumor, retinal vasculitis, ocular allergy, chronic conjunctivitis. Many of the above described diseases and inflammation disorders., cause an increase of WBC's expressing the activated conformation of LFA- ⁇ to migrate and congregate i the ocular region and
  • There is evidence that indicates 'retinal inflammation' contributes to the pathogenesis of diabetic retinopathy.
  • cytokines There are common sets of inflammatory cytokines that are unregulated in both the serum and vitreous and aqueous samples, in subjects with diabetic retinopathy, and these cytokines can have multiple interactions to impact, the pathogenesis of the disease.
  • the key inflammatory events involved in the blood retinal barrier (B B) alteration appear to be: (1) increased expression of endothelial adhesion molecules such as ICAM 1 , VCAM L PECAM- 1 , and P-selectm, (2) adhesion of leukocytes to the endothelium.
  • Sjogren's syndrome is an inflammatory disease that can affect many different parts of the body, hut most often affects the tear and saliva glands. Patients with this condition may notice irritation, a gritty feeling, or painful burning in the eyes.
  • Membrane bound ICAM-.1 is over expressed i the salivary glands (SG) of Sjogren's syndrome (SS) patients.
  • Dry eye occurs when the eye does not produce tears properly, or when the tears are not of the correct consistency and evaporate too quickly. Dry eye can be associated with inflammation of the surface of the eye, the lacrimal gland,, or the conjunctiva.
  • Conjunctivitis is an inflammation of the conjunctiva.
  • the conjunctiva is the thin, transparent membrane lining that covers the outer surface of the eye.
  • the conjunctiva is nourished by tiny blood vessels that are nearly invisible to the naked eye.
  • the conjunctiva also secretes mucus that moistens and lubricates the eye.
  • Conjunctivitis that persists for four or more weeks is considered chronic, Conjunctivitis can be caused by an infection or by an allergen.
  • allograft rejection is the main cause of graft failure in human corneal transplantation.
  • LFA-1 in conjunction with ICAM-J has been shown to be important in animal models of corneal graft rejection, thus allograft rejection is another condition that would be suitable for treatment by the methods of the present invention.
  • subject refers to human and non-human animals.
  • non-human animals include all vertebrates, e.g., mammals such as non-human primates (particularly higher primates), dogs, rodents (e.g., mice or rats), guinea pigs, cats and non-mammals, such as birds, amphibians, reptiles, etc.
  • the subject is a human.
  • the subject is an experimental animal or animal suitable as a disease model. (00491 A subject to be treated for ocular inflammatory disease can be identified b standard diagnosing techniques.
  • the subject can be examined for the level or percentage of W BCs that bind to LtxA in a test sample obtamed from the subject by methods described below. If the binding level or percentage is at or abo ve a threshold value (which can be obtained from a normal subject), the subject is a candidate for treatment with an effecti ve amount of LtxA.
  • a threshold value which can be obtained from a normal subject
  • Treating * ' or ''treatment refers to administration of a compound or pharmaceutical composition to a subject, who has an ocular inflammatory disease, with the purpose to cure, alleviate, relieve, remedy, delay the onset of. or ameitoraie the disorder, the symptom of the disorder, the disease state secondary to the disorder, or the predisposition toward the disorder.
  • a “therapeutically effective amount” refers to the amount of an agent or pharmaceutical composition sufficient to effect beneficial or desired results.
  • a therapeutically effective amount can be administered in one or more administrations, applications or dosages and is not intended to be i united to a particular formulation or administration route.
  • the LtxA pharmaceutical composition can be administered in vivo or ex vivo., alone or co-administered in conjunction with other drugs or therapy.
  • coadministration or “co-administered” refers to the administration of at least two agent(s) or therapies to a subject. In some embodiments, the co-administration of two or more agents/therapies is concurrent. In other embodiments, a first agent/therap is administered prior to a second agent/therapy.
  • the LtxA pharmaceutical composition can be administered local iy by topically applying the LtxA pharmaceutical composition to the eye in the form of an ointment, gel or droppable liquids using an ocular delivery system known to the art such as an applicator or eyedropper.
  • the LtxA may be administered intraocularly via a polymer implant that is placed tinder the under the conjunctiva of the eye or through injection directly into the eye.
  • a pharmaceutical composition comprising LtxA can be administered systemic-ally.
  • LtxA is suspended in a phatnraceuiically-aeceptable carrier (e.g., physiological saline) and administered orally or by intravenous infusion, or injected subcutaneously, intrarauscu!arly, intrathecaily, intra peritoneal ly, mtranasaUy, intragastrical y, mrratracheally. or mtrapulmonarily.
  • a phatnraceuiically-aeceptable carrier e.g., physiological saline
  • administered orally or by intravenous infusion or injected subcutaneously, intrarauscu!arly, intrathecaily, intra peritoneal ly, mtranasaUy, intragastrical y, mrratracheally. or mtrapulmonarily.
  • ⁇ 0QSSJ The dosage required depends on the choice of the route of administration; the nature of the formulation; the nature of the patient's illness; the subject's size, weight, surface area, age, and sex; other drags being administered; and the judgment of the attending physician. Suitable dosages are in the range of 0.01-100 mg kg. Variations in the needed dosage are to be expected in view of the variety of compounds available and the different efficiencies of various rouies of administration. Variations in these dosage levels can be adjusted using standard empirical routines for optimization as is wel! understood in the art that may be employed by the ordinarily skilled artisan without undue experimentation. Encapsulation of the compound in a suitable delivery vehicle (e.g., polymeric microparticles or implantable devices) may increase the efficiency of delivery.
  • a suitable delivery vehicle e.g., polymeric microparticles or implantable devices
  • Ocular inflammation can be characterized by an increase in active WBCs expressing a greater level of the activated conformation of LFA-I compared to WBC's of a healthy subject without ocular inflammation.
  • LFA- 1 present on WBCs of ocular inflammatory disease patients, can act as a marker to detect and monitor the treatment of these afflictions while providing a therapeutic target for pharmaceutical agents.
  • LtxA specifically targets WBCs that express the activated conformation of LFA-1., and therefore can be used in diagnosing diseases meditated by such WBCs.
  • the present invention provides a. method to diagnose ocular inflammation characierized by the increase of activated LFA-1 WBCs in. the ocular region.
  • WBCs expressing the ac tivated conformation of LFA-1 can be detected in. a subject based on the presence of the binding of LtxA in a test sample from the subject.
  • the binding of LtxA can be used as markers to indicate the presence or absence of WBCs involved in ocular inflammatory disease.
  • Diagnostic and prognostic assays of the invention include methods for assessing the binding level of LtxA with WBCs in the eye and the surrounding tissue.
  • the binding level in a test sample can be evaluated by obtaining a test sample from a test subject and contacting the test sample with LtxA.
  • the "test sample” includes tissues, cells and biological fluids isolated from a subject, as well as tissues., cells and fluids present within, a subject, and from, the ocular region.
  • the level of binding of LtxA to WBCs can be measured in a number of ways, including that described in the examples below.
  • LtxA or its fragments that mediate binding between LtxA and LFA-1 are labeled with a detectable agent.
  • labeled is intended to encompass direct labeling of the probe by physically linking a detectable substance to the probe, as well as indirect labeling of the probe by reactivit with a detectable substance.
  • LtxA (or its fragment) can be indirectly labeled using a second antibody directed against LtxA, wherein the second antibody is coupled to a detectable substance.
  • detectable substances or labels include radio isotopes (e.g., 1251, 1311, 35S, 3H, or 32P), enzymes (e.g., alkaline phosphatase, horseradish peroxidase, luciferase, or ⁇ -glactosidase), fluorescent moieties or proteins (e.g., fluorescein isothiocyanate. rhodaniine, phycoerythrin, GFP, or BFP), or luminescent moieties (e.g., QdotTM nanoparticles by the Quantum Dot Corporation, Palo Alto, CA).
  • radio isotopes e.g., 1251, 1311, 35S, 3H, or 32P
  • enzymes e.g., alkaline phosphatase, horseradish peroxidase, luciferase, or ⁇ -glactosidase
  • fluorescent moieties or proteins e.g., fluoresc
  • LtxA not only binds to, but also kills, WBCs.
  • the binding of LtxA. and WBCs can be conducted at low temperatures (e.g., 0-4°C) and for a short period of time such as 5 to 20 or 30 minutes.
  • the prognostic assays described herein can be used to determine whether a. subject is suitable to be administered with an agent (e.g., a drug) to treat an ocular inflammatory disease.
  • an agent e.g., a drug
  • such assays can be used to determine whether a subject can be administered with a cytotoxic drug or immune-suppressants to treat ocular inflammatory disease.
  • a biological sample from a subject can also be screened for the increased expression of certain cytokines (biomarkers) to determine whether the subject is in need of a treatment for ocular inflammation.
  • cytokines include IL-4, IL-5, 1L- 9, IL-17F and IL-23a.
  • Standard assays are known in the art to detect cytokines in biological samples, including without limitation biological samples isolated f om the ocular region.
  • the binding level between LtxA and WBCs can be determined for test samples from a subject before, during, or after undergoing a treatment.
  • a decrease in the binding level after the treatment indicates that the subject can be further treated by the same treatment.
  • a patient who has received a cornea transplant often faces the problem of tissue rejection . That is, the body has an immune response to a cornea transplant.
  • cornea transplant is often accompanied by nonspecific immune suppression therapy to prevent T cell-mediated rejection.
  • LtxA's binding level can serve as a marker for a proper level or degree of immune suppression, and/or biomarker expression level be determined compared to the normal cytokine level of a subject withotn ocular inflammation.
  • a person skilled in the art can adjust the amount of Lt A administered and lengt of treatment based on the level of the binding during the course of the treatment.
  • kits containing a pharmaceutical ly acceptable dose unit of a pharmaceutically effective amount of leukoioxin, and a set of instructions containing methods of treating an ocular inflammatory disorder as described.
  • the kit may further contain an applicator to administer the pharmaceutically acceptable dose unit of a pharmaceutically effective amount of leukoto i to the ocular region.
  • Ocular applicators are known in the art.
  • LtxA Leukotoxin
  • HBECs were grown to monolayer on collagen or gelatin and then stimulated with TNF.
  • Cakem-iabeied PBMCs activated with PMA were then added to the monolayer and incubated for 2 hours. Unbound cells were washed and the percent PBMC binding was calculated by measuring fluorescence.
  • LtxA was able to prevent binding of activated PBMCs to the endothelial cells in a dose-dependent manner. Greater than 50% blocking was observed at higher doses of LtxA. Because these PBMCs were from a healthy individual, complete blocking was not observed since most of the cells were normal and not affected by LtxA.
  • LtxA can block the migration of monocytes across a human brain endothelial cell layer (HBECs). LtxA has the ability to selectively deplete and block activated PBMCs from binding to and crossing endothelial cells.
  • Ltx A was next tested in the rhesus macaque (Macaca mulatta) under physiological, conditions (PEG. 3A-F).
  • One monkey received a single intravenous infusion of 300 ⁇ g LtxA (22 ⁇ ig/kg) while the second animal served as a control.
  • red blood cell (RBC), platelet (PLT), and hemoglobin (HGB) values remained unchanged in the LtxA-treated monkey and were similar to the ehicle-treated control animal.
  • the total white blood cell (WBC), Lymphocyte (LY ) and Neutrophil (NEU) count in the treated animal dropped markedly during the first several hours post-infusion and then increased. By 24 hours, counts recovered and long-term myelosuppression was not observed.
  • Soluble LtxA was incubated at room temperature for several days and then tested for bioactivity against a sensitive leukemia cell line (THP-1). LtxA retains full activity even after seven days at room temperature in solution. Similar results were obtained for numerous different preparations of LtxA.
  • Effector memory T-cells were highly susceptible to LtxA while central memory T-cells were minimally affected and naive ⁇ -cells were not affected. Inflammatory eye diseases, such as dry eye, the effector T-cells exert damaging effects through their functions of inducing overt inflammatory responses even in the absence of antigen in tissues. Regulatory T- cells (Tregs) were not depleted by LtxA. Tregs are an important component of the immune system that prevents excessive immune reactions and can help abrogate autoimmune disease. Thus, LtxA acts on relevant cell types in a highly specific manner, sparing healthy and immunosuppressive cells that are. not involved in disease. Evaluation of LtxA in a Lung inflammation Mouse Model ,
  • LtxA was purified from culture supematants of A, actinomycetemconutans strain 4500 (Diaz et aL, (2006) Microb Pathog 40, 8-55.)
  • mice Female BALB/e mice (6-8 weeks, 15-20 , Jackson Laboratories, Bar Harbor. ME) were housed under specific pathogen-free conditions and a 12-hour light/dark cycle with access to food and water. Under isofJurane anesthesia, mice were exposed to HDM extract (D. pteronyssinus; Greer Laboratories, Lenoir, NC) intranasal iy (25 g in 10 ⁇ . ⁇ of saline per nostril). Control (non-asthmatic) animals were administered an equal volume of saline alone (saline/no treatment, FiG . 4). The frequency of exposure for HDM saline was 5 days/week, for a total of 5 weeks. Power calculations from pilot studies indicated that four animals per group were sufficient to detect a significant difference between control and experimental groups.
  • HDM extract D. pteronyssinus
  • Greer Laboratories Lenoir, NC
  • Control (non-asthmatic) animals were administered an equal volume of
  • HDM-exposed mice were divided into 4 groups of 4 mice/group and treated with (!) dexamethasone vehicle, saline (HDM/Dex vehicle, FIG. 4A-B) subcutaneously (s.c.) once daily, five days per week, (2) dexamethasone, Sigma; 1.25 rag kg (HDM/Dex., FIG. 4) s.c. once daily, five days per week, (3) LtxA vehicle, buffer (MDM/LtxA vehicle, F G. 4) tntraperitoneally (ip.) ; three days per week, and (4) LtxA, 0.5 mg/kg (HDM LtxA, FIG. 4) Lp. s three days per week. HDM exposure was continued throughout the 3- week treatment period.
  • BAL fluid from each mouse was subjected to RBC lysis for 10 minutes at room temperature, followed by washing twice at 400 x g for 5 minutes and resuspending the ceil peilet in PBS. Total BAL fluid cell number was counted using a hemocytometer. Immunophenotypic analysis of BAL fluid cells was performed by antibody staining and flow cytometry, for each antibody stain, 106 cells were first blocked with Fc blocker (rat anti-mouse CD16/CD32, BD Biosciences, San Jose, CA) for 10 minutes at room temperature, followed by incubation with monoclonal antibodies at 4° C for 30 miiiutes and analysis on an LSR O flow cyrometer (BD Bioscienees).
  • Fc blocker rat anti-mouse CD16/CD32, BD Biosciences, San Jose, CA
  • CD3e T-ceSIs
  • CD45 B220 B-cells
  • Ly6G Ly6G
  • CC 3 neutraltrophils
  • CCR3, Ly6G ⁇ eosinophils MHC 11 (macrophages).
  • Relevant isotype controls were included with each experiment.
  • ⁇ 0G77J Blood from the mice was collected by ileac vein puncture immediately following the BAL fluid isolation. Blood was collected in anticoagulant tubes and centrifused at 400 x a for 10 minutes.
  • Lung tissue histology After collecting BAL fluid and blood, lungs were removed through dissection and stored in 10% neutral buffered formalin at room temperature until microscopic analysis. Sections of paraffin embedded fixed king tissues were stained with H&E to analyze total lung inflammation. Sections were also stained with periodic acid Schiff reagent to identify mucous and goblet cell hyperplasia and Sirius red for eosinophils. The tissue preparation and examination was carried out at the New Jersey Medical School Histology Core Facility. Samples were examined by a board certified pathologist,
  • Cytokine Analysis Quantitative RT-PCR was used to determine the expression levels of proinflammatory cytokines (IL-4, ! L ⁇ 5, IL-9, IL-17F and lL ⁇ 23a) in the lungs of mice.
  • Total RNA from the lung tissue was extracted with Trizol. reagent (Life Technologies, Grand island. NY). Relative mRNA levels were determined by qRT-PC .
  • One microgram of total RN A was reverse transcribed using High Capacity cDNA Reverse Transcription Kit (Life Technologies, Grand Island, NY). Amplification was carried out using Taq an Fast Universal PC Master Mix (Life Technologies, Grand Island, NY). The data was normalized to GAPDB, Gene expression was calculated using the ⁇ method, relative to naive sample,
  • HDM-exposed mice were subdivided into four groups of four mice per group and received the following treatments for an additional three weeks: dexamethasone vehicle, subcutaneous (s,c.) 5 days/week; dexamethasone (1 ,25 mg kg), s,c. 5 days/week; LtxA vehicle, intraperitoneal (ip.) 3 days/week; LtxA (0.5 mg/kg), Lp, 3 days/week.
  • H&E staining revealed a large infiltration of WBCs in the long tissue of HDM-exposed mice treated with dexamethasone vehicle or LtxA vehicle, infiltration was not evident in saline-exposed mice.
  • the infiltration of WBCs in HDM-exposed mice was most evident surrounding the blood vessels and bronchioles.
  • Staining of polysaccharides with PAS in the lung tissue from LtxA vehicle-treated mice confirmed the presence of mucin- producing goblet ceils and subepithelial accumulation of collagen.
  • IL-4, IL-5, IL-9, IL-I7F, and IL-23a are the primary signaling molecules involved in disease.
  • the levels of IL-4, IL-5, IL-9, IL- I 7F, and IL-23a raRNA in the lung tissue from all mice were evaluated (FIG. 9).
  • the vehicle-treated mice had significantly greater expression of the proinflammatory cytokines compared to saline- exposed mice.
  • dexamethasone caused reduction of IL-9, 1 L- 17F. and iL-23a while LtxA treatment caused significant reduction of all the cytokines that were examined.

Abstract

This invention concerns compositions and methods of treating or diagnosing ocular disease.

Description

TREATMENT AND DIAGNOSIS OF OCULAR DISEASE CROSS-REFERENCE TO RELATED APPLICATION
[000! J This application claims priority of U.S. Provisional Application No, 61/840,045 filed on June 27, 20.13. The content of the application is incorporated herein by reference in its entirety.
FIELD Of THE INVENTIO
[0002} This invention relates to pharmaceutical compositions, reagents and methods for treating or diagnosing ocular disease.
BACKGROUND OF THE INVENTION
|OO03J Ocular inflammatory disease is a general term embracing a number of diseases and conditions in whic inflammation affects the eye or surrounding tissues. Inflammation is due to abnormal activation and migration of white blood cells to regions for example., the conjunctival epithelium and tear glands. Although some treatments are presently available, many possess significant side effects or are not very effective in alleviating all symptoms. Thus, there is a need for drugs and reagents for treatment and diagnosis of ocular inflammatory disease.
SUMMARY OF THE INVENTION
|0Oo4) This invention relates to treatment and diagnosis of ocular inflammatory disease using leukotoxin (LfxA), a bacterial protein.
jOQOSJ At least one aspect of the invention is directed to a method of treating a disease characterized by ocular inflammation, the method comprising administering a pharmaceutical composition to a subject in need of such treatment i an amount effective to reduce said inflammation, wherein the pharmaceutical composition comprises a leukotoxin and a pharmaceutically acceptable carrier, and wherein the disease is an ocular inflammatory disease. The ocular inflammatory disease maybe keratoconjuuctivitis sicca, diabetic retinopath .. diabetic macula edema, Sjogren syndrome, dry eye, scleritis, birdshot retinochoriodopathy, ocular cicatricial pemphigoidm, keratitis, sympathetic ophthalmia, vogt-koyanagi harada, fuehs' helerochromk iridocyclitis, uveitis, pars planitis, episcleritis, optic neuritis, orbital pseudotumor, retinal vasculitis, ocular allergy, and chronic conjunctivitis.
{0006} Another aspect of the invention is directed to method of reducing ocular inflammation in an ocular regio within a subject in need thereof, characterized by increased levels of activated white blood cells in a subject in need thereof, the method comprising administering to the subject an amount of a pharmaceutical composition in amount effective to reduce said ocular inflammation, wherein the pharmaceutical composition comprises a leukotoxin and a pharmaceutically acceptable carrier. The ocular inflammation may be caused by an infection., autoimmune disease, allergen, ocular trauma, and ocular allograft rejection. The infection may be a viral, bacterial, or a fungal infection.
[00071 1ft at least oae embodiment, the leukotoxin is LtxA, prepared from Aggregatibacter actinomyceiemcomitans. In yet another embodiment, LtxA has ai least 90% homology with the peptide according to SEQ ID NO: ί .
[00081 1ft another embodiment, the pharmaceutical composition comprising leukotoxin is administered topically to the ocular region. In another embodiment, the pharmaceutical composition is in a form suitable for topical administration. In another embodiment, the pharmaceutical composition is formulated for and administered by using an eye drop dispenser, in another embodiment, the pharmaceutically acceptable carrier is an eye gel, eye cream, suspension-type eye drops, eye wash, ointment, gel, liposomal dispersion, colloidal microparticle suspension, implant or a contact lens. |ftO 9| In another embodiment the anio tit of the pharmaceutical composition comprising leukotoxin administered to a subject in need thereof is effective to reduce local cytokine levels in the ocular region. One or more of the following cytokines expression levels are reduced, iL-4, IL~5 JL~9; ILHF and IL~23a.
|001OJ Another aspect of the invention is directed to a kit comprising a pharmaceutically effective amount of leukotoxin for the treatment of an ocular inflammatory disease. The kit may contain multiple doses of a pharmaceutical composition comprising m effective amount of leukotoxin to treat an ocular inflammatory disease or to reduce ocular inftammation characterized by the increase of activated white blood ceils in the ocular region that express a greater level of LFA- 1 compared to white blood cells from a normal healthy subject.
DESCRIPTION OF DRAWINGS
{001 If FIG. I is a diagram showing migration of white blood ceils (WBCs) across an endothel ial barrier.
(00! 21 FIG. 2 is a diagram showing sensitivity of activated T-ceils,
(6013) FIG. 3A-F is a set of diagrams showing the activity of LtxA under physiologic conditions.
(001 ) FIG. 4 is a diagram showing the examination of cytokines in Sung tissue following treatment of house dust mite (HDM) exposed mice with either a vehicle (saline, HDM/Dex vehicle; or HDM/LtxA vehicle); dexaroethasone (HDM/Dex); or leukotoxin (HDM/LtxA), DETAILED DESCRIPTION OF THE INVENTIO
(90.15) This invention relates to reagents and methods for treating or diagnosing ocular inflammatory disease and incorporates the discovery that administering LxA to a subject sufferin ocular inflammation characterized by activaied inflammatory cells expressing LFA-1 results in the depletion of the activated inflammatory cells.
(00161 LFA-1 , a $2-integrin on the surface of white blood cells, is composed of CD! la and CD! 8 and involved in immune cell migration and signaling. in the absence of infection, circulating WBCs express a "resting state" LFA-1 on their surface. Inflammatory cytokines cause LFA-1 to assume an active conformation, which results in binding of activated LFA-1 to intercellular adhesion molecule- 1 (iCAM-1 ) on the surface of endothelial cells. The interaction between LFA-1 and CAM-l results in migration of WBCs across the endothelial barrier and into the infected tissue, it has now been discovered that LtxA targets inflammatory white blood cells WBCs thai express the activated conformation of LFA-1 o their surface that migrate to the eye and surrounding tissue resulting i ocular inflammation and local organ damage.
{(1017| Ocular inflammatory disease is a general term for inflammation affecting the ocular region. The ocular region means any part of the eye or surrounding tissue, as known by one with ordinary skill in the art. Inflammation involving the eye can range from the familiar allergic conjunctivitis of hay fever to rare, potentially blinding conditions such as uveitis, scleiitis, episcleritis, optic neuritis, keratitis, orbital pseudotumor, retinal vasculitis, and chronic conjunctivitis.
fOOISJ Broadly speaking, if inflammation develops in the eye(s), or in the optic nerve, blood vessels, muscles or other tissues that surround the eye, the resulting illness is referred to as an ocular inflammatory disease.
|0O19| The location of the inflammation may govern the diagnostic name for the ocular inflammatory disease. For example, uveitis is inflammation in the uveal tract; sc!eritis is inflammation of the sclera, pars planitis is inflammation of the pars plana, and so forth. The treatment of ocular inflammatory disease is aimed at .reducing inflammation. Ocular inflammation can occur for many reasons for example, as a resul t of an infec tion, an autoimmune disorder, trauma to the eye, or allergies.
LtxA
10020} LtxA is a - 1 15 kDa protein produced by the Gram negative bacterium Aggregatibacter actinomycetemeomitans (Kac any, S. C. 2010. J Dent Res 89:563-570.). LtxA specifically kills leukocytes of humans and Old World Primates by forming pores in the membrane and causing apoptosis or necrosis (Mangan et a!., 1991. infect Immim 59:3267-72.). LtxA binds specifically to LFA~i and cells that lack LFA-1 are resistant to its toxicity (KachJany, S. C. et al., 2010. Leukemia Research 34:777-85.}, For example, LtxA is not. active against human red. blood ceils, human epithelial cells, rat cells, or mou.se cells. LtxA also remains active in the presence of human peripheral blood.
10021} Many LtxA preparations can be used, highly purified LtxA is preferred. Examples include LtxA polypeptide purified from Aggregatibacter actinomycetemeomitans (SEQ ID NO: 1 shown above) and other variants having substantially the same bioiogical activity as that having the sequence of SEQ ID NO: 1. it was discovered that Aggregatibacter actinomycetemeomitans secreted active LtxA into culture superaatants (Kaeh'iany, S. C, et al. 2000. infect Immun 68:6094-100) and an efficient method for its purification was described in Kach!any, S. C, et at 2002. Protein Expr Purif 25:465-71. This method can therefore be used to prepare isolated or purified LtxA polypeptide. In one example, a purification procedure of the toxin involves;
a. inoculating a single colony of Aggregatibacter actinomycetemeomitans into a fresh broth and growing cultures; b. adding the growing cultures to fresh broth, adding glass beads and incubating;
c. centrifuging the incubated culture, forming a pellet and a supernatant:
d. filtering the supernatant through a membrane to provide a filtered supernatant;
e. .mixing (NH4)2S04 and the filtered supernatant together to form a mixture;
f. centrifuging the mixture to form, a mixture pellet;
g. resuspending the mixture pellet in buffer to form a protein resuspension;
h. passing the protein resuspension through a column; and
i. collecting the protein e ting off the column.
See also PCI7US2006/45258 (WO 2007/062150); US Application 20090075883 (US Set. No. 12/154,843) and PCX/ US 10/52453 (WO 201 1 /04701 1). The contents of these documents are incorporated herein by reference.
0 22| An "isolated polypeptide" refers to polypeptide that has been separated from other proteins, lipids, and nucleic acids with which it is naturally associated. The term "polypeptide" and "protein" can be used interchangeably when referring to LtxA. The polypeptide constitutes at least 10% (i.e., any percentage between 10% and 100%, e.g., 20%, 30%. 40%, 50%, 60%, 70 %, 80%, 85%, 90%, 95%, and 99%) by dry weight of the purified preparation. Purity can be measured by any appropriate standard method, for example, by column chromatography, polyacrylaxnide gel electrophoresis, or HPLC analysis. An isolated polypeptide of the invention can be purified from a natural source, produced by recombinant DNA techniques, or by chemical methods. A functional equivalent of LtxA refers to a polypeptide derivative of the LtxA polypeptide, e.g., a protein having one or more point mutations, insertions, deletions, truncations, a fusion protein, or a combination thereof. It retains substantially the activity of the LtxA polypeptide, i.e., the ability to .target and kill WBCs that express the activated conformation of LFA~1. on their surface while having littie or no toxic effect o» other cells or organs in the body. The isolated polypeptide can contain SEQ I D NO; i or a functional fragment of SEQ ID NO; L In general, the functional equivalent is at least 75% (e.g., any number between 75% and 100%, inclusive, e.g., 70 %, 80%, 85%, 90%, 95%, and 99%) identical to SEQ ID NO: I .
|0023| All of naturally occurring LtxA, genetic engineered LtxA, and chemically synthesized LtxA can be used to practice the invention disclosed herein. LtxA obtained by recombinant NA technology may have the same amino acid sequence as naturally a occurring LtxA (SEQ 3D NO: 1 ) or an functionally equivalent thereof. The term "LtxA" also covers chemically modified LtxA. Examples of chemically modified LtxA include LtxA subjected to conformational change, addition or deletion of a sugar chain, and LtxA to which a compound such as polyethylene glycol has been bound. Once purified and tested by standard methods or according to the method described in the examples below. LtxA can be included in a pharmaceutical composition, e.g., a topical composition.
(00241 The amino acid composition of the LtxA polypeptide described herein may vary without disrupting the ability of the polypeptide to target and kill WBCs. For example, it can contain one or more conservative amino acid substitutions. A "conservative amino acid substitution" is one in which the amino acid residue is replaced with an amino acid residue having a similar side chain. Families of amino acid residues having similar side chains have been defined in the art. These families include amino acids with basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g., glycine, asparagine, glutaniine, serine, threonine, tyrosine, cysteine), nonpolar side chains (e.g., alanine, valine, leucine, isoleueine, proline, phenylalanine, methionine, tryptophan), beta-branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g.. tyrosine, phenylalanine, tryptophan, histidine). Thus, a predicted nonessential amino acid residue in SEQ ID NO; 1 is preferably replaced with another amino acid residue from the same side chain family. Alternatively, mutations can be introduced randomly along all or part of SEQ ID NO: , such as by saturation mutagenesis, and the resultant mutants can be screened for the ability to mprove the ocular condition and/or to identify mutants that retain, the activity as described below in the examples.
[0025} A LtxA polypeptide as described in this invention can be obtained as a naturally occurring polypeptide or a recombinant polypeptide. To prepare a recombinant, polypeptide, a nucleic acid encoding it (e.g., SEQ ID NO: 2) can be linked to another nucleic acid encoding a fusion partner., e.g., gl«tathione-s-transferase (GST), 6x-His epitope tag, or l 3 Gene 3 protein. The resultant fusion nucleic acid expresses in suitable host cells a fusion protein that can be isolated by .methods known in the art. The isolated fusion protein can be further treated, e.g. , by enzymatic digestion, to remove the fusion partner and obtain the recombinant polypeptide of this invention.
Pharmaceutical Compositions
|0026| The present invention also provides a pharmaceutical composition that contains LtxA and a pharmaceutically acceptable carrier suitable for ocular administration. Examples of pharmaceutically acceptable carriers include, but are not limited to eye gel, eye cream, suspension-rype eye drops, eye wash, ointment, gel, liposomal dispersion, colloidal microparticle suspension, contact lens, and the like, and other preparations known in the art to be suitable for ocular administration. As such, the pharmaceutical compositions of the present invention containing LtxA may be admimstered using commonly known devices configured for the delivery of the pharmaceutical compositions in the form of to the region surrounding the eye. An ocular insert, may also comprise a biodegradable controlled release polymeric matrix. The ocular insert can be implanted in the conjunctiva, sclera, pars plana, anterior segment, or posterior segment of the eye. Implants provide for controlled release of the pharmaceutical composition to the ocular surface, typically sustain, release of LtxA over an extended time period. The pharmaceutically acceptable carrier of the pharmaceutical composition of the invention may comprise a wide variety of non-active ingredients which are useful for formulation purposes and which do not materially affect the novel and useful properties of LtxA.
{0G27J By a "pharmaceutically acceptable" or "ophthalmologically acceptable" component, means a component that is not biologically or otherwise undesirable, i.e., the component may be incorporated into a pharmaceutical composition of the invention and administered topically to a patient's eye without causing any undesirable biological effects or interacting in a deleterious manner with any of the other components of the formulation composition in which it. is contained. When the teen "pharmaceutically acceptable" is used to refer to a component other than a pharmacologically active agent, it is implied that the component has met the required standards of toxicological and manufacturing testing or that it is included on the Inactive Ingredient Guide prepared by the U.S. Food and Drug Administration.
|tMl28| Suitable thickeners will be known to those of ordinary skill in the art of ophthalmic formulation, and include, by way of example, cellulosic polymers such as methyl cellulose (MC), hydroxyethyketlulose (HEC), hvdroxypropylcellubse {HPC), hydrox ropylmeth Ice 1.1 nlose (HPMC), and sodium carboxyraethylce!luJose ( aCMC), and other swellabte hydrophilic polymers such as polyvinyl alcohol (PVA), hyaluronic acid or a salt thereof (e.g., sodium hyaluronate), and cross!inked acrylic acid polymers commonly referred to as "carboraers". The preferred amount of any thickener is such that a viscosity in the range of about 15 cps to 25 cps is provided, as a solution having a viscosity in the aforementioned range is generally considered optimal for both comfort and retention of the formulation in the eye. Any suitable isotonic agents and buffering agents commonly used in ophthalmic formulations may be used, providing that the osmotic pressure of the solution does not deviate from that of lachrimal fluid by more than 2-3% and that the pH of the .formulation is maintained in. the range of about 6.5 to about 8.0, preferably in the range of about 6.8 to about 7.8, and optimally at a pH of about 7.4. Preferred buffering agents include carbonates such as sodium and potassium bicarbonate.
|0029| The pharmaceutical compositions of the invention may also be prepared as a hydrogei, dispersion, or colloidal suspension. Hydrogels are formed by incorporation of a swei iahie, gel- forming polymer such as those set forth above as suitable thickening agents (i.e., MC, EEC, HPC, HPMC, NaCMC, PVA, or hyaluronic acid or a salt thereof, e.g., sodium hyaluronate), except that a formulation referred to in the ait as a "hydrogei"' typically has a higher viscosity than a formulation referred to as a "thickened" solution or suspension. In contrast to such preformed hydrogels, a pharmaceutical composition may also be prepared so as to form a hydrogei in situ following application to the eye. Such gels are liquid at room temperature but get at higher temperatures (and thus are termed "thermoreversible" hydrogels), such as when placed in contact with body fluids. Biocompatible polymers thai impart this property include acrylic acid polymers and copolymers, N-isopropylacrylamide derivatives, and ABA block copolymers of ethylene oxide and propylene oxide. The pharmaceutical compositions can also be prepared in the form of a dispersion or colloidal suspension. Preferred dispersions are liposomal, in which case the pharmaceutical composition is enclosed within "liposomes," microscopic vesicles composed of alternating aqueous compartments and lipid biiayers. Colloidal suspensions are generally formed from micropariicles, i.e., from microspheres. nanospheres, microcapsules, or nanocapsules, wherein microspheres and nanospheres are generally monolithic particles of a polymer matrix in which the pharmaceutical composition is trapped, adsorbed, or otherwise contained, while with microcapsules and nanocapsules, the formulation is actually encapsulated.
|0030| The pharmaceutically acceptable ophthalmic carrier used with the pharmaceutical composition of the invention may be of a wide range of types known to those of skill in the art. For example, the pharmaceutical compositions of the invention can be provided as an ophthalmic solution or suspension, in which case the carrier is at least partially aqueous. The pharmaceutical compositions may also be ointments, in which case the pharmaceutical ly acceptable carrier comprises an ointment base. Preferred ointment bases herein have a melting or softening point close to body temperature, and any ointment bases commonly used in ophthalmic preparations ma be advantageously employed. Common ointment bases include petrolatum and mixtures of petrolatum and mineral oil.
(00311 The pharmaceutical composition may also be incorporated into a sterile ocular insert tha provides for controlled release of the fonnulation over an extended time period, generally in the range of about 12 hours to 60 days, and possibly up to 12 months or more, following implantation of the insert into the conjunctiva, sclera, or pars plana, or into the anterior segment or posterior segment of the eye. One type of ocular insert is an implant in the form of a monolithic polymer matrix that, gradually releases the pharmaceutical composition to the eye through diffusion and/or matrix degradation. With such an insert, it is preferred that the polymer be completely soluble and or biodegradable (i.e., physically or enzymatically eroded in tire eye) so that removal of the insert is unnecessary. These types of inserts are well known in the art, and are typically composed of a water-swellable, gel-forming polymer such as poly ethylene glycol. collagen, polyvinyl alcohol, or a cellulosic polymer. Another type of insert thai can be used to deliver the present formulation is a diffusional implant in which the formulation is contained in a central reservoir enclosed within a permeable polymer membrane that allows for gradual diffusion of the pharmaceutical composition out of the implant. Osmotic inserts may also he used, i.e., implants in which the pharmaceutical composition is released as a result of an increase in osmotic pressure within the implant following application to the eye and subsequent absorption of tears.
|0032| The term "controlled release" refers to an agent-containing foundation or fraction thereof in which release of the agent is not immediate, i.e., with a "controlled release" formulation, administration does not result in immediate release of the agent into an absorption pool The term is used interchangeably with "nonimmediate release" as defined in Remington: The Science and Practice of Pharmacy, Nineteenth Ed. (Fasten, Pa.: Mack Publishing Company, 1995). in general, the term "controlled release" as used herein refers to "sustained release" rather than to "delayed release" formulations. The terra "sustained release" (synonymous with "extended release") is used in its conventional sense to refer to a formulation that provides for gradual release of an agent over an extended period of time.
|«033) In an embodiment, the LtxA is released over a period of at least 12 hours, at least 18 hours, at least 24 hours, at least 48 hours, at least 3 days, at least 7 days, at least 14 days, at least 30 days, at least 60 days, at least 90 days, at least 100 days, at least 120 days, at least 150 days, at least 180 days, or even longer.
|0O34| The LtxA or pharmaceutical composition can be administered, as described herein, according to any of a number of standard methods including, but not limited to injection, nasal spray, time-release implant, transdermal patch, eye drops, gels, ointments, orally, intraocular injection, subeonjuetival injection, peri-Zreirobulbar injection, transdermal^, or topically to tlie ocular region by an eye drop dispenser, or the like, including topical intranasal administration or administration by inhalant, and the like, spray, emulsion, suspension, via any drug carriers as sponges, contact lenses, polymers, microspheres, and implants. The topical administration can he ophihalmically or intraaasa!ly.
f0035| Topical ophthalmic products may be packaged in multidose form. Preservatives are thus required to prevent microbial contamination during use. Suitable preservatives include: biguanides, hydrogen peroxide, hydrogen peroxide producers, ben alkonium chloride, chlorobutanol, benzododectniu bromide, methyl paraben, propyl parahen, phenylethyl alcohol, edetate disodium, sorbic acid, po!yqiiatemium-3 , or other agents known to those skilled in the art. Such preservatives are typicall employed at a level of from 0.001 to 1 % (w/w). Unit, dose formulations of the present invention will, be sterile, but typically unpreserved. Such formulations, therefore, generally will not contain preservatives.
(00361 The pharmaceutical composition may further comprise antibiotics. Examples of antibiotics include without limitation, cefazolin, cephradine, cefaclor, cephapirin, ceftizoxime, cefoperazone, cefotetan, cefutoxime, cefotaxime, cefadroxil, ceftazidime, cephalexin, cephalothin, cefamandole, cefoxitin, cefonieid, cefbranide, ceftriaxone, cefadroxil, cephradine, cefuroxime, ampicillin, amoxicillin, cyelacillm, ampicillin, penicillin G, penicillin V potassium, piperacillin, oxacillin, bacampicillin, cioxacillin, ticarci!ltn, azlociiUn. carbenicillin, methicilHn, nafcillin, erythromycin, tetracycline, doxycycline, minocycline, aztreonam, chloramphenicol, ciprofloxacin hydrochloride, clindamycin, metronidazole, gentamtcin, lincomycin, tobramycin, vancomycin, polymyxin B sulfate, colistimethate, eolistin, azithromycin, augraentin, sulfamethoxazole, trimethoprim, derivatives thereof, and. the like and mixtures thereof. |ft037| The pharmaceutical composition may further comprise corticosteroids. Examples of corticosteroids include cortisone, prednisolone, triamcinolone, fiaromethokm\ dexameihasone, medrysone, loteprednol, fiuazacort, hydrocortisone, prednisone triamcinolone, betamethasone, prednisone, methyl prednisolone, triamcinolone acetoni.de, triamcinolone hexacatonide, parame hasone aceiate, difiorasone. fluocinolooe and fluoctnonide, derivatives thereof, and mixtures thereof.
[9038) The pharmaceutical composition may further comprise immunosuppressive agents. Examples of immunosuppresive agents include cyclosporine, azathioprine, tacrolimus, T Fa- inhibitors. infliximab, (Remicade), adalimumah Humira), ceriolizumab pegol (Cimzia), and golimumab (Stmponi), and etanereept (Enbrei) and derivatives thereof.
[0039] The pharmaceutical composition ma further comprise antiviral agents. Examples of antiviral agents include and are not limited to, interferon gamma, zidovudine, amantadine hydrochloride, ribavirin, acyclovir, valciclovir, dideoxycytidaie, and derivatives thereof.
(00401 The pharmaceutical composition may further comprise antihistamines. Examples of antihistamines include, and are not limited to, loradatine, hydroxyzine, diphenhydramine, chlorpheniramine, brompheniramine, cyproheptadine, terfenadine, clemastine, triprolidine, carbinox amine, diphenylpyr aline, phenindamine, azatadine, tripe lennamine, dexchlorpheniraniine, dexbrompheniramine, methdilaziiie, and trimprazine dox lamine, pheniramine, pyri!amine, chiorcycliztne, thonzylamine, and derivatives thereof
Treatmen t Methods
(0041 J The invention provides a method of treating a disease characterized by ocular inflammation in a subject characterized by increased le vels of activated WBCs, by administering to the subject a pharmaceutical composition comprising LtxA and a pharmaceutically acceptable carrier in an amount effective to reduce ocular inflammation characterizing the disease. In another embodiment, the invention provides a method of reducing ocular inflammation in a subject characterized by increased levels of activated WBCs, by administering to the subject a pharmaceutical composition comprising leufcotoxm and a pharmaceutically acceptable carrie in an amount effective to reduce ocular inflammation.
|9Θ42| Ocular inflammatory disease is characterized by a number of diseases and conditions in which inflammation affects the eye or surrounding tissues. Examples of ocular inflammatory disease include keratoconjunctivitis sicca, diabetic retinopathy, diabetic macula edema, Sjogren syndrome, dry eye, scleritis, birdshot retinochoriodopathy, ocular cicatricial pemphigoid, keratitis, sympathetic ophthalmia, vogt-koyanagi liarada, fuchs* beterochromic iridocyclitis- uveitis, pars piarsitis, episcleritis, optic neuritis, orbital pseudotumor, retinal vasculitis, ocular allergy, chronic conjunctivitis. Many of the above described diseases and inflammation disorders., cause an increase of WBC's expressing the activated conformation of LFA-Ϊ to migrate and congregate i the ocular region and are suitable for treatment by the methods of the present invention.
{0043| There is evidence that indicates 'retinal inflammation' contributes to the pathogenesis of diabetic retinopathy. There are common sets of inflammatory cytokines that are unregulated in both the serum and vitreous and aqueous samples, in subjects with diabetic retinopathy, and these cytokines can have multiple interactions to impact, the pathogenesis of the disease. The key inflammatory events involved in the blood retinal barrier (B B) alteration appear to be: (1) increased expression of endothelial adhesion molecules such as ICAM 1 , VCAM L PECAM- 1 , and P-selectm, (2) adhesion of leukocytes to the endothelium. (3) release of inflammatory chemokioes, cytokines, and vascular permeability factors, (4) alteration of adherens and tight junctional proteins between the endothelial cells, and (5) infiltration of leukocytes into the neuro- retina, resulting in the alteration of the blood retinal barrier (diapedesis).
[004 j Sjogren's syndrome is an inflammatory disease that can affect many different parts of the body, hut most often affects the tear and saliva glands. Patients with this condition may notice irritation, a gritty feeling, or painful burning in the eyes. Membrane bound ICAM-.1 is over expressed i the salivary glands (SG) of Sjogren's syndrome (SS) patients.
[9045) Dry eye occurs when the eye does not produce tears properly, or when the tears are not of the correct consistency and evaporate too quickly. Dry eye can be associated with inflammation of the surface of the eye, the lacrimal gland,, or the conjunctiva.
[0046] Conjunctivitis is an inflammation of the conjunctiva. The conjunctiva is the thin, transparent membrane lining that covers the outer surface of the eye. The conjunctiva is nourished by tiny blood vessels that are nearly invisible to the naked eye. The conjunctiva also secretes mucus that moistens and lubricates the eye. Conjunctivitis that persists for four or more weeks is considered chronic, Conjunctivitis can be caused by an infection or by an allergen.
[0047) Another condition, allograft rejection is the main cause of graft failure in human corneal transplantation. LFA-1 in conjunction with ICAM-J has been shown to be important in animal models of corneal graft rejection, thus allograft rejection is another condition that would be suitable for treatment by the methods of the present invention.
(0048J A "subject" refers to human and non-human animals. Examples of non-human animals include all vertebrates, e.g., mammals such as non-human primates (particularly higher primates), dogs, rodents (e.g., mice or rats), guinea pigs, cats and non-mammals, such as birds, amphibians, reptiles, etc. In a preferred embodiment, the subject is a human. In another embodiment, the subject is an experimental animal or animal suitable as a disease model. (00491 A subject to be treated for ocular inflammatory disease can be identified b standard diagnosing techniques. Optionally, the subject can be examined for the level or percentage of W BCs that bind to LtxA in a test sample obtamed from the subject by methods described below. If the binding level or percentage is at or abo ve a threshold value (which can be obtained from a normal subject), the subject is a candidate for treatment with an effecti ve amount of LtxA.
(00501 "Treating*' or ''treatment" refers to administration of a compound or pharmaceutical composition to a subject, who has an ocular inflammatory disease, with the purpose to cure, alleviate, relieve, remedy, delay the onset of. or ameitoraie the disorder, the symptom of the disorder, the disease state secondary to the disorder, or the predisposition toward the disorder.
[0051 J A "therapeutically effective amount" refers to the amount of an agent or pharmaceutical composition sufficient to effect beneficial or desired results. A therapeutically effective amount can be administered in one or more administrations, applications or dosages and is not intended to be i united to a particular formulation or administration route.
(00521 The LtxA pharmaceutical composition can be administered in vivo or ex vivo., alone or co-administered in conjunction with other drugs or therapy. As used herein, the term "coadministration" or "co-administered" refers to the administration of at least two agent(s) or therapies to a subject. In some embodiments, the co-administration of two or more agents/therapies is concurrent. In other embodiments, a first agent/therap is administered prior to a second agent/therapy. Those of skill m the art understand, mat the formulations and/or routes of administration of the various agents/therapies used may vary.
{0053J The LtxA pharmaceutical composition can be administered local iy by topically applying the LtxA pharmaceutical composition to the eye in the form of an ointment, gel or droppable liquids using an ocular delivery system known to the art such as an applicator or eyedropper. Alternatively, the LtxA may be administered intraocularly via a polymer implant that is placed tinder the under the conjunctiva of the eye or through injection directly into the eye. {"0054} In another embodiment, a pharmaceutical composition comprising LtxA can be administered systemic-ally. Generally, LtxA is suspended in a phatnraceuiically-aeceptable carrier (e.g., physiological saline) and administered orally or by intravenous infusion, or injected subcutaneously, intrarauscu!arly, intrathecaily, intra peritoneal ly, mtranasaUy, intragastrical y, mrratracheally. or mtrapulmonarily.
{0QSSJ The dosage required depends on the choice of the route of administration; the nature of the formulation; the nature of the patient's illness; the subject's size, weight, surface area, age, and sex; other drags being administered; and the judgment of the attending physician. Suitable dosages are in the range of 0.01-100 mg kg. Variations in the needed dosage are to be expected in view of the variety of compounds available and the different efficiencies of various rouies of administration. Variations in these dosage levels can be adjusted using standard empirical routines for optimization as is wel! understood in the art that may be employed by the ordinarily skilled artisan without undue experimentation. Encapsulation of the compound in a suitable delivery vehicle (e.g., polymeric microparticles or implantable devices) may increase the efficiency of delivery.
Diagnostic and Prognostic Methods
{0056} Ocular inflammation can be characterized by an increase in active WBCs expressing a greater level of the activated conformation of LFA-I compared to WBC's of a healthy subject without ocular inflammation. LFA- 1 , present on WBCs of ocular inflammatory disease patients, can act as a marker to detect and monitor the treatment of these afflictions while providing a therapeutic target for pharmaceutical agents. LtxA specifically targets WBCs that express the activated conformation of LFA-1., and therefore can be used in diagnosing diseases meditated by such WBCs.
{"0057] In another embodiment the present invention provides a. method to diagnose ocular inflammation characierized by the increase of activated LFA-1 WBCs in. the ocular region. WBCs expressing the ac tivated conformation of LFA-1 can be detected in. a subject based on the presence of the binding of LtxA in a test sample from the subject. In other words, the binding of LtxA can be used as markers to indicate the presence or absence of WBCs involved in ocular inflammatory disease. Diagnostic and prognostic assays of the invention include methods for assessing the binding level of LtxA with WBCs in the eye and the surrounding tissue.
[0058] The binding level in a test sample can be evaluated by obtaining a test sample from a test subject and contacting the test sample with LtxA. The "test sample" includes tissues, cells and biological fluids isolated from a subject, as well as tissues., cells and fluids present within, a subject, and from, the ocular region. The level of binding of LtxA to WBCs can be measured in a number of ways, including that described in the examples below. In a preferred embodiment, LtxA or its fragments that mediate binding between LtxA and LFA-1 (i.e., probes) are labeled with a detectable agent. The term "labeled" is intended to encompass direct labeling of the probe by physically linking a detectable substance to the probe, as well as indirect labeling of the probe by reactivit with a detectable substance. For example, LtxA (or its fragment) can be indirectly labeled using a second antibody directed against LtxA, wherein the second antibody is coupled to a detectable substance. Examples of detectable substances or labels include radio isotopes (e.g., 1251, 1311, 35S, 3H, or 32P), enzymes (e.g., alkaline phosphatase, horseradish peroxidase, luciferase, or β-glactosidase), fluorescent moieties or proteins (e.g., fluorescein isothiocyanate. rhodaniine, phycoerythrin, GFP, or BFP), or luminescent moieties (e.g., QdotTM nanoparticles by the Quantum Dot Corporation, Palo Alto, CA).
{"0059} LtxA not only binds to, but also kills, WBCs, In the diagnostic or prognostic method, to minimize any potential, errors caused by cell death, the binding of LtxA. and WBCs can be conducted at low temperatures (e.g., 0-4°C) and for a short period of time such as 5 to 20 or 30 minutes.
[0060} The prognostic assays described herein can be used to determine whether a. subject is suitable to be administered with an agent (e.g., a drug) to treat an ocular inflammatory disease. For example, such assays can be used to determine whether a subject can be administered with a cytotoxic drug or immune-suppressants to treat ocular inflammatory disease.
(00611 In another embodiment, a biological sample from a subject can also be screened for the increased expression of certain cytokines (biomarkers) to determine whether the subject is in need of a treatment for ocular inflammation. These cytokmes biotnarkers include IL-4, IL-5, 1L- 9, IL-17F and IL-23a. Standard assays are known in the art to detect cytokines in biological samples, including without limitation biological samples isolated f om the ocular region.
{0062} Thus, also 'featured in this invention is a method of monitoring a treatment for ocular inflammatory disease in a subject. For this purpose, the binding level between LtxA and WBCs can be determined for test samples from a subject before, during, or after undergoing a treatment. A decrease in the binding level after the treatment indicates that the subject can be further treated by the same treatment. For example, a patient who has received a cornea transplant often faces the problem of tissue rejection . That is, the body has an immune response to a cornea transplant. To address this problem, cornea transplant is often accompanied by nonspecific immune suppression therapy to prevent T cell-mediated rejection. To that end, LtxA's binding level, can serve as a marker for a proper level or degree of immune suppression, and/or biomarker expression level be determined compared to the normal cytokine level of a subject withotn ocular inflammation. A person skilled in the art can adjust the amount of Lt A administered and lengt of treatment based on the level of the binding during the course of the treatment.
|0063| Information obtained from the practice of the above assays is useful in prognostication, identifying progression of, and clinical management of diseases and other deleterious conditions affecting an individual's health status. The information more specifically assists the clinician in designing therapy or other treatment regimens for ocular inflammatory disease.
Kits
|0064| in another aspect, the invention provides kits. In one embodiment, a kit containing a pharmaceutical ly acceptable dose unit of a pharmaceutically effective amount of leukoioxin, and a set of instructions containing methods of treating an ocular inflammatory disorder as described. The kit may further contain an applicator to administer the pharmaceutically acceptable dose unit of a pharmaceutically effective amount of leukoto i to the ocular region. Ocular applicators are known in the art.
EXAMPLE
f0065| Leukotoxin (LtxA) kills WBCs and. prevents migration of WBCs across an endothelial barrier even at doses that do not kill cells (FIG. I ).
1006i>| Testing to determine if LtxA could block binding of acti vated PBMCs to human brain endothelial ceils (HBECs) to model the initial steps leading to inflammation were carried out. The initial steps leading to inflammation include the binding of activated PBMCs to endothelial cells and migration across the endothelial barrier. Strategies to treat inflammatory response include depletion of those WBCs that are involved in pathogenesis and interruption of their binding and migration into the affected tissue. (Weksler, B. B., E. A. Subileau, N, Perriere, P. Charaeau, , Holloway, M. Leveque, H. Trscoire-Leignel. A, Nicofra, S, Bourdoulous, P. Turowski, D. K. Male. F. R ux, J. Greenwood,, I. A. Romero, and P. O. Couraud.. 2005, Blood- brain barrier-specific properties of a human adult brain endothelial cell line. Faseb J 19:1872-4.) or (Wassmer, S. C, V. Combes, F. J. Caudal, I. Juhan- Vague, and G. E. Grau. 2006. Platelets potentiate brain endothelial alterations induced by Plasmodium falciparum. Infect Imtmm 74:645-53.)
{(KM»7| HBECs were grown to monolayer on collagen or gelatin and then stimulated with TNF. Cakem-iabeied PBMCs activated with PMA were then added to the monolayer and incubated for 2 hours. Unbound cells were washed and the percent PBMC binding was calculated by measuring fluorescence. LtxA was able to prevent binding of activated PBMCs to the endothelial cells in a dose-dependent manner. Greater than 50% blocking was observed at higher doses of LtxA. Because these PBMCs were from a healthy individual, complete blocking was not observed since most of the cells were normal and not affected by LtxA. Cytotoxicity assays showed that only approximately 10-20% of the cells stained with propidium iodide after two hours thereby indicating that many of the cells that were blocked by LtxA were not killed. LtxA can block the migration of monocytes across a human brain endothelial cell layer (HBECs). LtxA has the ability to selectively deplete and block activated PBMCs from binding to and crossing endothelial cells.
j0068j Next, the specificity of LtxA by examining the effects of LtxA on activated versus non-activated (resting) WBCs was demonstrated. Primary CD4 T-celis were stimulated by cross-linking their T~celi receptor with anti~CD3 CD28 mAb. which is known to cause physiological activation- We then tested the effects of LtxA on activated CD4 T~eeils. The activated CD4 T-cells were more than 100-times more sensitive to LtxA than resting T-cells (FIG. 2), Healthy PBMCs were minimally affected by LtxA while the same PBMCs activated ex vivo with I2-0>tetradecanoylphorboH 3-ac-e.tate (TPA) were highly sensitive to LtxA -mediated killing. Analysis of LFA-1 on activated ceils revealed that LtxA preferentially depletes cells with the highest level of LFA-1 activation as revealed by binding to mAb24, which binds specifically to LFA-1 in the activated state, but not in the resting state.
|¾069] Ltx A was next tested in the rhesus macaque (Macaca mulatta) under physiological, conditions (PEG. 3A-F). One monkey received a single intravenous infusion of 300 μg LtxA (22 ^ig/kg) while the second animal served as a control. Throughout the 3-week study, red blood cell (RBC), platelet (PLT), and hemoglobin (HGB) values remained unchanged in the LtxA-treated monkey and were similar to the ehicle-treated control animal. In contrast, the total white blood cell (WBC), Lymphocyte (LY ) and Neutrophil (NEU) count in the treated animal, dropped markedly during the first several hours post-infusion and then increased. By 24 hours, counts recovered and long-term myelosuppression was not observed.
{0070| Blood chemistry values for the LtxA-treated monkey revealed no change in markers for either liver toxicity (AST, bi lirubin, alkaline phosphatase) or kidney function (BUN, creatinine) during the course of the experiment, and animals appeared in good health as evaluated twice daily by the veterinary staff (data not shown). Taken together, these results indicate that LtxA is active and specific in a non-human primate at a non-toxic dose. Primate experiments were carried out at the University of Wisconsin National Primate Research Center (WNPRC) and approved by the University of Wisconsin-Madison Animal Care and Use Committee. |ft07!| LtxA in solution is highly stable. Soluble LtxA was incubated at room temperature for several days and then tested for bioactivity against a sensitive leukemia cell line (THP-1). LtxA retains full activity even after seven days at room temperature in solution. Similar results were obtained for numerous different preparations of LtxA. We also tested the stability of LtxA at different temperatures. LtxA was incubated for ten minutes at 40, 50, 60, 70, 80, and 90° C and then tested against THP-1 cells. LtxA was stable up to 60° C and then began to lose activity. We tested the stability of LtxA. at 40* C and 50c C over time and found, that LtxA retains full activity even after a 4-hour incubation at 40° C or 50° C. LtxA is also fully stabl after ten r eeze-thaw cycles.
[Θ072| Immunophenotypic analysis of WBCs was performed in rhesus monkeys. LtxA (22 g/kg) was infused i.v. into two monkeys over a twenty-minute period and then blood was drawn from animals at different time points. WBCs were analyzed by flow cytometry. T-cells (CD3+), B-cells (CD20-;-), and natural killer (N'K) ceils were depleted by LtxA. N cells, which have bee shown to have an important role in the pathogenesis of inflammatory eye diseases, demonstrated the greatest sensitivity to LtxA. Only the cells with highest levels of LFA-1 were targeted by LtxA. Effector memory T-cells were highly susceptible to LtxA while central memory T-cells were minimally affected and naive Γ-cells were not affected. Inflammatory eye diseases, such as dry eye, the effector T-cells exert damaging effects through their functions of inducing overt inflammatory responses even in the absence of antigen in tissues. Regulatory T- cells (Tregs) were not depleted by LtxA. Tregs are an important component of the immune system that prevents excessive immune reactions and can help abrogate autoimmune disease. Thus, LtxA acts on relevant cell types in a highly specific manner, sparing healthy and immunosuppressive cells that are. not involved in disease. Evaluation of LtxA in a Lung inflammation Mouse Model,
Materials and Methods
{"0073} LtxA Purification; LtxA was purified from culture supematants of A, actinomycetemconutans strain 4500 (Diaz et aL, (2006) Microb Pathog 40, 8-55.)
{"0074} Animal study. Female BALB/e mice (6-8 weeks, 15-20 , Jackson Laboratories, Bar Harbor. ME) were housed under specific pathogen-free conditions and a 12-hour light/dark cycle with access to food and water. Under isofJurane anesthesia, mice were exposed to HDM extract (D. pteronyssinus; Greer Laboratories, Lenoir, NC) intranasal iy (25 g in 10 μ.Ι of saline per nostril). Control (non-asthmatic) animals were administered an equal volume of saline alone (saline/no treatment, FiG . 4). The frequency of exposure for HDM saline was 5 days/week, for a total of 5 weeks. Power calculations from pilot studies indicated that four animals per group were sufficient to detect a significant difference between control and experimental groups.
10075} After two weeks, HDM-exposed mice were divided into 4 groups of 4 mice/group and treated with (!) dexamethasone vehicle, saline (HDM/Dex vehicle, FIG. 4A-B) subcutaneously (s.c.) once daily, five days per week, (2) dexamethasone, Sigma; 1.25 rag kg (HDM/Dex., FIG. 4) s.c. once daily, five days per week, (3) LtxA vehicle, buffer (MDM/LtxA vehicle, F G. 4) tntraperitoneally (ip.); three days per week, and (4) LtxA, 0.5 mg/kg (HDM LtxA, FIG. 4) Lp.s three days per week. HDM exposure was continued throughout the 3- week treatment period.
[00761 BAL fluid from each mouse was subjected to RBC lysis for 10 minutes at room temperature, followed by washing twice at 400 x g for 5 minutes and resuspending the ceil peilet in PBS. Total BAL fluid cell number was counted using a hemocytometer. Immunophenotypic analysis of BAL fluid cells was performed by antibody staining and flow cytometry, for each antibody stain, 106 cells were first blocked with Fc blocker (rat anti-mouse CD16/CD32, BD Biosciences, San Jose, CA) for 10 minutes at room temperature, followed by incubation with monoclonal antibodies at 4° C for 30 miiiutes and analysis on an LSR O flow cyrometer (BD Bioscienees). Antibodies (BD Biosciences) to the following markers were used to identify the various cellular subtypes: CD3e (T-ceSIs). CD45 B220 (B-cells), Ly6G, CC 3 (neutrophils), CCR3, Ly6G {eosinophils), MHC 11 (macrophages). Relevant isotype controls were included with each experiment.
{0G77J Blood from the mice was collected by ileac vein puncture immediately following the BAL fluid isolation. Blood was collected in anticoagulant tubes and centrifused at 400 x a for 10 minutes.
j00?8| Lung tissue histology: After collecting BAL fluid and blood, lungs were removed through dissection and stored in 10% neutral buffered formalin at room temperature until microscopic analysis. Sections of paraffin embedded fixed king tissues were stained with H&E to analyze total lung inflammation. Sections were also stained with periodic acid Schiff reagent to identify mucous and goblet cell hyperplasia and Sirius red for eosinophils. The tissue preparation and examination was carried out at the New Jersey Medical School Histology Core Facility. Samples were examined by a board certified pathologist,
[0079.1 Cytokine Analysis: Quantitative RT-PCR was used to determine the expression levels of proinflammatory cytokines (IL-4, ! L~5, IL-9, IL-17F and lL~23a) in the lungs of mice. Total RNA from the lung tissue was extracted with Trizol. reagent (Life Technologies, Grand island. NY). Relative mRNA levels were determined by qRT-PC . One microgram of total RN A was reverse transcribed using High Capacity cDNA Reverse Transcription Kit (Life Technologies, Grand Island, NY). Amplification was carried out using Taq an Fast Universal PC Master Mix (Life Technologies, Grand Island, NY). The data was normalized to GAPDB, Gene expression was calculated using the ΔΔΟΤ method, relative to naive sample,
{"0080} Evaluation of LtxA in a mouse model for allergic asthma. Given the potential role that LFA-1 plays in the pathogenesis of allergic asthma and the ability .for LtxA to target specifically the LFA- lhi WBCs ex vivo that are unique to allergic asthma patients, an initial proof-of-pr ciple evaluation of LtxA in a mouse model for allergic asthma was performed. Mice were administered house dust mite (HDM) extract or saline intranasally (in.) five days per week for five weeks. After two weeks of administration, HDM-exposed mice were subdivided into four groups of four mice per group and received the following treatments for an additional three weeks: dexamethasone vehicle, subcutaneous (s,c.) 5 days/week; dexamethasone (1 ,25 mg kg), s,c. 5 days/week; LtxA vehicle, intraperitoneal (ip.) 3 days/week; LtxA (0.5 mg/kg), Lp, 3 days/week.
1 081} At the end of the study;, bronchoalveo'lar lavage (BAL) fluid, lung tissue, and blood were collected from all mice for further evaluation. Examination of WBCs in the BAL fluid revealed, that HDM-exposed mice treated with the dexamethasone vehicle or LtxA vehicle had significantly higher levels of all WBC subsets than mice that were given only saline. Treatment of HDM-exposed mice with dexamethasone or LtxA caused significant reduction in the numbers of WBCs in the BAL fluid.
(0082 J To determine if LFA- 1 is involved in the migration of WBCs to the lung tissue in this animal model, the levels of LFA-1 on P'BMCs and BAL fluid WBCs in two HDM-exposed mice that were treated with LtxA vehicle were examined. The migrated WBCs that were present in the BAL fluid had significantly higher levels of LFA-1 than on the WBCs in the peripheral blood o the same animal. |ft083| Lung tissue was sectioned and stained with H&E, PAS, or Sirius Red. H&E staining revealed a large infiltration of WBCs in the long tissue of HDM-exposed mice treated with dexamethasone vehicle or LtxA vehicle, infiltration was not evident in saline-exposed mice. The infiltration of WBCs in HDM-exposed mice was most evident surrounding the blood vessels and bronchioles. Significant goblet cell hyperplasia surrounding many of the bronchioles in the vehicle-treated controls, bat not in the other samples was observed. Staining of polysaccharides with PAS in the lung tissue from LtxA vehicle-treated mice confirmed the presence of mucin- producing goblet ceils and subepithelial accumulation of collagen. Sirius Red staining of sections revealed pink-staining eosinophils in the vehicle-treated mice, but not in. the LtxA- treated mice. Mice that were treated with dexamethasone had a reduced, number of eosinophils compared, to the vehicle control, but still greater than LtxA-treated mice.
ffl084| Proinflammatory cytokines pla a crucial role in the pathogenesis of allergic asthma and other inflammatory conditions, in allergic asthma, lL-4, l'L-5, IL-9, IL-I7F, and IL-23a are the primary signaling molecules involved in disease. The levels of IL-4, IL-5, IL-9, IL- I 7F, and IL-23a raRNA in the lung tissue from all mice were evaluated (FIG. 9). The vehicle-treated mice had significantly greater expression of the proinflammatory cytokines compared to saline- exposed mice. In addition, dexamethasone caused reduction of IL-9, 1 L- 17F. and iL-23a while LtxA treatment caused significant reduction of all the cytokines that were examined.
{"0085 j AH publications cited in the specification publications, are indicative of the level of skill of those reasonably skilled in the art to which this invention pertains. Al l these publications ate herein fully incorporated by reference to the same extent as if each individual publication were specifically and individually indicated as being incorporated by reference.

Claims

What is claimed is:
.! . A method of treating a disease characterized by ocular inflammation, the method comprising administering a pharmaceutical composition to a subject in need of such treatment in an amount effective to reduce said inflammation, wherein the pharmaceutical composition comprises a Ieukotoxin and a pharmaceutically acceptable carrier, and wherein the disease is an ocular inflammatory disease,
2, A method of reducing ocular inflammation in an ocular region within a subject in need thereof characterized by increased, levels of activated white blood cells in a subject in need thereof, the method comprising administering to the subject an amount of a pharmaceutical composition in amount effective to reduce said ocular inflammation, wherein the pharmaceutical composition comprises a ieukotoxin and a phamiaceutically acceptable carrier.
3, The method of claim 1 or 2, wherein the Ieukotoxin is prepared from Aggregatibacter actinotnycetemcomitans.
4. The method of claim I or 2, wherein the Ieukotoxin has at least 90% homology with the peptide according to SEQ ID NO; I .
5. The method of claim 1. wherein the ocular inflammatory disease is selected from the group consisting of keratoconjunctivitis sicca, diabetic retinopathy, diabetic macula edema, Sjogren syndrome, dry eye, scleritis, birdshot retmochoriodopathy, ocular cicatricial pempliigoidm, keratitis, sympathetic ophthalmia, vogt-koyanagi harada, fucbs' heterochromic iridocyclitis;, uveitis, pars plamtis, episcleritis, optic neuritis, orbital pseudotumor, retinal vasculitis, ocular allergy, and chroni conjunctivitises. The method of claim I or 2, wherein the pharmaceutical composition is administered topically to the ocular region. ?. The method of claim 1 or 2, wherem the pharmaceutical composition is administered
Figure imgf000031_0001
8. The method of claim I or 2, wherein the amount administered to said subject is effective to reduce local cytokine levels in the ocular region.
9. The method of claim 7 or 8, wherem one or more cytokines are selected from the group consisting of IL-4, IL-5, IL-9, IL17F and IL-23CL
10. The method of claim 1 or 2, wherein the pharmaceutical composition is formulated for and administered by using an eye drop dispenser,
1 L The method of claim 2, wherein the activated white blood cells express a greater level of LFA-1 compared to white blood cells from a normal healthy subject.
12. The method of claim 2, wherein the inflammation is caused by a disease or conditio selected from the group consisting of an infection, autoimmune disease, allergen, ocular trauma, and ocular allograft rejection.
13. The method of claim 12, wherein the infection may be caused by a virus, bacteria, or a fungus.
14. A pharmaceutical composition, comprising a leukotoxin and a pharmaceutical carrier, wherein the pharmaceutical composition is in a form suitable for topical administration,
15. The pharmaceutical composition of claim 14, wherei the pharmaceutical carrier is selected from the grou consisting of an eye gel, eye cream, suspension-type eye drops, eye wash, ointment, gel, liposomal dispersion, colloidal microparticle suspension, implant and a contact lens.
16. The pharmaceutical composition of claim 35, wherein the pharmaceutical carrier is a controlled release formulation.
17. Leukotoxin LtxA (SEQ ID NO:.l) or a polypeptide having at least 90% identity with SEQ ID NO: 1 and retaining the activity of the LtxA polypeptide for use in treating an ocular inflammatory disease .
18. Leukotoxin LtxA or polypeptide for us according to claim 17, wherein, the ocular inflammatory disease is selected from the group consisting of keratoconjunctivitis sicca, diabetic retinopathy, diabetic macula edema, Sjogren syndrome, dry eye, scleritis, hir shot reimo toriodopathy, ocular cicatricial pemphigoid.!), keratitis, sympathetic ophthalmia, vogi- koyanagi harada, fuchs' heterochromic iridocyclitis, uveitis, pars plamtis, episcleritis, optic neuritis, orbital pseudotumor, retinal vasculitis., ocular allergy, infection, and chronic conjunctivitis.
19. Use of Leukotoxin LtxA (SEQ ID NO: I) or a polypeptide having at least 90% identity with. SEQ ID NO: I for the maniiiacture of a medicament for treating an ocular inflammatory disease.
20. A kit comprising a pharmaceutically acceptable dose unit of a pharmaceutically effective amount of leukotoxin for the treatment of an ocular inflammatory disease.
PCT/US2014/044567 2013-06-27 2014-06-27 Treatment and diagnosis of ocular disease WO2014210454A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
JP2016524239A JP6544806B2 (en) 2013-06-27 2014-06-27 Treatment and diagnosis of eye disease
US14/901,631 US9950030B2 (en) 2013-06-27 2014-06-27 Treatment and diagnosis of ocular disease
CA2917056A CA2917056C (en) 2013-06-27 2014-06-27 Treatment and diagnosis of ocular disease
EP14817083.0A EP3013425B1 (en) 2013-06-27 2014-06-27 Leukotoxin for use in the treatment of ocular disease

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201361840045P 2013-06-27 2013-06-27
US61/840,045 2013-06-27

Publications (1)

Publication Number Publication Date
WO2014210454A1 true WO2014210454A1 (en) 2014-12-31

Family

ID=52142714

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2014/044567 WO2014210454A1 (en) 2013-06-27 2014-06-27 Treatment and diagnosis of ocular disease

Country Status (5)

Country Link
US (1) US9950030B2 (en)
EP (1) EP3013425B1 (en)
JP (1) JP6544806B2 (en)
CA (1) CA2917056C (en)
WO (1) WO2014210454A1 (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3157542A4 (en) * 2014-06-20 2018-02-07 Rutgers, the State University of New Jersey Methods to treat inflammation of the lung
US10149889B2 (en) 2005-11-25 2018-12-11 Rutgers, The State University Of New Jersey Compositions for the treatment of cancer, and methods for testing and using the same
US10226506B2 (en) 2009-10-13 2019-03-12 Rutgers, The State University Of New Jersey Treatment and diagnosis of inflammatory disorders and HIV
JP2019506840A (en) * 2015-11-27 2019-03-14 ヴィラマティクス エスディーエヌ ビーエイチディー Peptides as antiviral agents and uses therefor
US11376306B2 (en) 2017-05-26 2022-07-05 Viramatix Sdn Bhd Peptides and uses therefor as antiviral agents

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20190388548A1 (en) * 2018-06-26 2019-12-26 Tzu Chi University Method for providing ocular neuroprotection or for preventing, treating or alleviating the effects of, an ocular disease associated with retinal ganglion cell death

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060252107A1 (en) * 2005-02-22 2006-11-09 Acucela, Inc. Compositions and methods for diagnosing and treating retinal diseases
WO2007062150A2 (en) 2005-11-25 2007-05-31 University Of Medicine And Dentistry Of New Jersey Leukotoxin compositions and therapeutic methods
US20090075883A1 (en) 2005-11-25 2009-03-19 Kachlany Charles S Compositions for the treatment of cancer, and methods for testing and using the same
WO2011047011A2 (en) 2009-10-13 2011-04-21 University Of Medicine And Dentistry Of New Jersey Treatment and diagnosis of inflammatory disorders
US20120263644A1 (en) * 2009-10-13 2012-10-18 University Of Medicine And Dentistry Of New Jersey Treatment and diagnosis of inflammatory disorders and hiv

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004090095A2 (en) * 2003-04-01 2004-10-21 University Of Southern California Generation of human regulatory t cells by bacterial toxins for the treatment of inflammatory disorders
EP1943350A4 (en) * 2005-10-04 2009-06-17 Greenville Hospital System Latent procytotoxins and uses thereof
US9352017B2 (en) * 2011-03-16 2016-05-31 Rutgers, The State University Of New Jersey Combination therapy with leukotoxin

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060252107A1 (en) * 2005-02-22 2006-11-09 Acucela, Inc. Compositions and methods for diagnosing and treating retinal diseases
WO2007062150A2 (en) 2005-11-25 2007-05-31 University Of Medicine And Dentistry Of New Jersey Leukotoxin compositions and therapeutic methods
US20090075883A1 (en) 2005-11-25 2009-03-19 Kachlany Charles S Compositions for the treatment of cancer, and methods for testing and using the same
WO2011047011A2 (en) 2009-10-13 2011-04-21 University Of Medicine And Dentistry Of New Jersey Treatment and diagnosis of inflammatory disorders
US20120263644A1 (en) * 2009-10-13 2012-10-18 University Of Medicine And Dentistry Of New Jersey Treatment and diagnosis of inflammatory disorders and hiv

Non-Patent Citations (9)

* Cited by examiner, † Cited by third party
Title
"Remington: The Science and Practice of Pharmacy", 1995, MACK PUBLISHING COMPANY
DIAZ ET AL., MICROB PATHOG, vol. 40, 2006, pages 48 - 55
KACHLANY, S. C. ET AL., INFECT IMMUN, vol. 68, 2000, pages 6094 - 100
KACHLANY, S. C. ET AL., LEUKEMIA RESEARCH, vol. 34, 2010, pages 777 - 85
KACHLANY, S. C. ET AL., PROTEIN EXPR PURIF, vol. 25, 2002, pages 465 - 71
KACHLANY, S. C., J DENT RES, vol. 89, 2010, pages 561 - 570
MANGAN ET AL., INFECT IMMUN, vol. 59, 1991, pages 3267 - 72
WASSMER, S. C.V. COMBESF. J. CANDALI. JUHAN-VAGUEG. E. GRAU: "Platelets potentiate brain endothelial alterations induced by Plasmodium falciparum", INFECT IMMUN, vol. 74, 2006, pages 645 - 53
WEKSLER, B. B.E. A. SUBILEAUN. PERRIEREP. CHARNEAUK. HOLLOWAYM. LEVEQUEH. TRICOIRE-LEIGNELA. NICOTRAS. BOURDOULOUSP. TUROWSKI: "Blood-brain barrier-specific properties of a human adult brain endothelial cell line", FASEB J, vol. 19, 2005, pages 1872 - 4, XP002658008, DOI: 10.1096/fj.04-3458fje

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10149889B2 (en) 2005-11-25 2018-12-11 Rutgers, The State University Of New Jersey Compositions for the treatment of cancer, and methods for testing and using the same
US10226506B2 (en) 2009-10-13 2019-03-12 Rutgers, The State University Of New Jersey Treatment and diagnosis of inflammatory disorders and HIV
US10603354B2 (en) 2009-10-13 2020-03-31 Rutgers, The State University Of New Jersey Treatment and diagnosis of inflammatory disorders and HIV
US10993989B2 (en) 2009-10-13 2021-05-04 Rutgers, The State University Of New Jersey Treatment and diagnosis of inflammatory disorders and HIV
US11612636B2 (en) 2009-10-13 2023-03-28 Rutgers, The State University Of New Jersey Method of treating autoimmune inflammatory crohn's disease
EP3157542A4 (en) * 2014-06-20 2018-02-07 Rutgers, the State University of New Jersey Methods to treat inflammation of the lung
JP2019506840A (en) * 2015-11-27 2019-03-14 ヴィラマティクス エスディーエヌ ビーエイチディー Peptides as antiviral agents and uses therefor
US11376306B2 (en) 2017-05-26 2022-07-05 Viramatix Sdn Bhd Peptides and uses therefor as antiviral agents

Also Published As

Publication number Publication date
JP2016523913A (en) 2016-08-12
US20160287662A1 (en) 2016-10-06
US9950030B2 (en) 2018-04-24
JP6544806B2 (en) 2019-07-17
CA2917056A1 (en) 2014-12-31
CA2917056C (en) 2020-03-24
EP3013425B1 (en) 2020-09-09
EP3013425A1 (en) 2016-05-04
EP3013425A4 (en) 2017-01-04

Similar Documents

Publication Publication Date Title
US9950030B2 (en) Treatment and diagnosis of ocular disease
Brooks et al. Diseases of the cornea
EP3429557A1 (en) Methods of treating ocular inflammation and chemical injuries of the eye with extracellular vesicles
Sadrai et al. Effect of topical azithromycin on corneal innate immune responses
KR20150083117A (en) Methods and devices for the treatment of ocular disease in human subjects
US8043609B2 (en) Viral complement control proteins for eye disorders
CN106668852B (en) Composition for treating and/or preventing type I diabetes and application thereof
AU2008289552A1 (en) Therapeutic compositions for treatment of inflammation of ocular and adnexal tissues
WO2006042252A2 (en) Viral complement control proteins for eye disorders
US9498517B2 (en) Stanniocalcin-1 (STC-1) therapy for treatment of retinal diseases
US20210386837A1 (en) Active low molecular weight variants of angiotensin converting enzyme 2 (ace2) for the treatment of diseases and conditions of the eye
JP2016527250A (en) Ophthalmic use of toxin-based therapeutic peptides and pharmaceutical compositions thereof
US20180236035A1 (en) Protein therapy for treatment of retinal diseases
WO2016060916A1 (en) Histatins as therapeutic agents for ocular surface disease
WO2017062491A1 (en) Therapeutic compositions for the treatment of dry eye and related ocular surface diseases
WO2011097577A2 (en) Compositions and methods for treating or preventing retinal degeneration
RU2564912C2 (en) Method of agent screening, suitable for treatment of dry eye syndrome and/or injury of cornea and conjunctiva, pharmaceutical composition, obtained by thereof
US20220034903A1 (en) Therapies That Target Autoimmunity For Treating Glaucoma And Optic Neuropathy
US20190247302A1 (en) Materials and methods for treating ophthalmic inflammation
CN112569338B (en) Application of TDFA in preparation of medicine for preventing and/or treating ocular surface inflammatory diseases
WO2018027149A1 (en) Methods of treating alport syndrome
US20220288028A1 (en) A therapeutic approach for treating non-infectious ocular immunoinflammatory disorders
WO2021163304A1 (en) Activation of neuropeptide receptors on plasmacytoid dendritic cells to treat or prevent ocular diseases associated with neovascularization and inflammation
Shah Evaluation of Rapamycin for the Treatment of Autoimmune-Mediated Dry Eye in a Mouse Model of Sjögren's Syndrome
CN114224910A (en) Application of fucoidan in preparation of medicine for treating prostatitis

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 14817083

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2016524239

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 14901631

Country of ref document: US

ENP Entry into the national phase

Ref document number: 2917056

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2014817083

Country of ref document: EP